Skip to main content

Full text of "USPTO Patents Application 08442277"

See other formats



t* <r C 1 C O S~ 



: ^3tP--N£OTTRTAiri4E^T^ STEM 



AND PROGENITOR CELLS OF THE BLOOD 



TABLE OF CONTENTS 



Page 



1Q 1. Introduction , 6 

2. Background of the Invention 6 

2.1. Hematopoietic Stem and Progenitor Cells.... 6 

2.2. Reconstitution of the Hematopoietic 

System 9 

15 2.3. Cryopreservation of Cells 13 

2.4. Gene Therapy 14 

3. Summary of the Invention 16 

3.1. Definitions 16 

4. Description of the Figures. 19 

2Q 5. Detailed Description of the Invention 20 

5.1. Isolation of Fetal or Neonatal 

Hematopoietic Stem and Progenitor Cells.... 25 

5.1.1. Collection of Neonatal Blood 26 

5,1.1.1. Volume 26 

2 5 5.1.1.2. Preferred Aspects 28 

5.1.1.2.1. Collection Kit 28 

5.1.1.2.2. Vaginal Delivery of the 

Term Infant 29 

5.1.1.2.3. Other Circumstances of 

3 q Birth and Delivery 30 

5.1.1.2.3.1. Premature Birth 3 0 

5.1.1.2.3.2. Multiple Births 30 

5.1.1.2.3.3. Caesarian Delivery.. 31 

5.1.1.2.3.4. Complicated Delivery 31 

35 




-2- 



Page 

5. 1.1.2. 3.5. Abnormal Placenta... 3 2 

5.1.1.2.3.6. Collection from the 
Delivered Placenta.. 3 2 

5 

5.1.1.2.3.7. Medical Conditions 

of the Mother 3 2 

5.1.1.2.3.8. Unplanned Delivery.. 3 3 
5.1.1.2.4. Recordation of Data 3 3 

5.1.2. Inspection and Testing of Neonatal 

10 Blood 34 

5.1.3. Optional Procedures 3 6 

5.1.3.1. Enrichment for Hematopoietic 
Stem and Progenitor Cells: 

Cell Separation Procedures.... 37 

15 

5.1.3.2. In Vitro Cultures of Hemato- 
poietic Stem and Progenitor 

Cells 43 

5.2. Cryopreservation 4 3 

5.3. Recovering Stem and Progenitor Cells from 

zu the Frozen State 4 7 

5.3.1. Thawing 4 7 

5.3.2. Optional Procedures 4 7 

5.4. Examination of Cells Recovered for 

Clinical Therapy 48 

25 5.4.1. Identity Testing 49 

5.4.2. Assays for Stem and Progenitor 

Cells 49 

5.5. Hematopoietic Reconstitution 50 

5.6. Therapeutic Uses 51 

30 - 

5.6.1. Diseases Resulting from a Failure 

or Dysfunction of Normal Blood 

Cell Production and Maturation 56 

5.6.2. Hematopoietic Malignancies 59 

35 

v3 



• -■• 

-3- 

Paqe 

5.6.3. Malignant Solid Tumors of Non- 
Hematopoietic Origin 59 

5.6.4. Autoimmune Disorders / 60 

5 

5.6.5. Gene Therapy 61 

5.6.6. Miscellaneous Disorders Involving 
Immune Mechanisms 64 

5.7 Generation and Use of Hematopoietic Stem 

and Progenitor Cell Progeny 65 

6. Examples 66 

6.1. Collection of Human Umbilical Cord Blood 

and Placental Blood 66 

6.2. Hematopoietic Stem and Progenitor Cells in 
Collected Cord Blood 68 

15 

6.3. Enrichment for Human Hematopoietic Stem 
and Progenitor Cells: Cell Separation 
Procedures 75 

6.3.1. Density Separations 76 

6.3.2. Adherence/Non-Adherence Separation. 80 

20 

6.4. Cryopreservation of Cord Blood Stem and 
Progenitor Cells 81 

6.5. Cell Thawing 82 

6.6. Human Hematopoietic Stem and Progenitor 

Cell Assays 8 3 

25 

6.6.1. CFU-GM Assay 8 4 

6.6.1.1. Preparation of McCoy's 

5 A Medium 86 

6.6.1.2. Preparation of Human 5637 
Urinary Bladder Carcinoma 

30 

Cell Line Conditioned 

Medium 8 7 

6.6.1.3. Preparation of Murine 
Pokeweed Mitogen Spleen 

Cell Conditioned Medium. . 88 

35 

4 




-4- 



Page 

6.6.2. BFU-E-2 and BFU-E-1/CFU-GEMM Assay.. 89 
6.6.2.1. Preparation of 2.1% Methyl 



Cellulose 91 

5 

6.6.2.2. Preparation of Hemin 92 

6.6.2.3. Preparation of Iscove's 
Modified Dulbecco's 

Medium 92 

6.6.3. Stem Cell Colony Forming Unit 

10 

Assay 93 

6.6.4. Assay of the Proliferative Status 

of Stem and Progenitor Cells 9 3 

6.7. Recovery After Freeze-Thawing of Human 
Hematopoietic Progenitor Cells 

15 

Derived from Cord Blood 9 5 

6.8. Calculations of the Reconstituting Potential 

of Cord Blood 98 

6.9. In Vitro Culture Conditions for Hematopoietic 
Stem and Progenitor Cells 100 

20 

6.10. Mouse Dissection Protocols 100 

6. 10.1. Bone Marrow Dissection 101 

6.10.2. Spleen Dissection 102 

6.11. Hematopoietic Reconstitution of Adult 
Mice with Syngeneic Fetal or Neonatal 

25 Stem Cells 103 

6.11.1. Hematopoietic Reconstitution 
of Lethally-Irradiated Mice 
with Stem Cells in Blood of the 

Near-Term Fetus 103 

30 .... 

6.11.2. Hematopoietic Reconstitution 

of Mice with a Lesser Volume of 

Near-Term Fetal Blood But Not 

with Adult Blood 105 

35 




-5- 



10 



Page 

6.11.3. Hematopoietic Reconstitution 
with Blood of Newborn Mice in 
Volumes as Low as Ten Microliters.. 108 

6.11.4. Hematopoietic Reconstitution 
with Blood of Newborn Mice in 
Volumes of 10 or 15 Microliters.... 110 

6.12 Hematopoietic Reconstitution For Treatment 

of Fanconi's Anemia Ill 

6.13. Flowchart: Description of a Service 114 



This applicat i^n^^ a Continuation - in - part of 



fob** 



15 



copendin g U.S. — application Serial No. — 07/119 , 746 -? — £iied A f , <s 0i 
N o vember 12, — 1 9 0 -7- , which is incorporated by reference herein in 
its entirety. **eLd 



20 



25 



30 



35 



-6- 



10 



15 



20 



25 



30 



35 



1. INTRODUCTION 

The present invention is directed to hematopoietic stem 
and progenitor cells of neonatal or fetal blood, that are 
cryopreserved, and the therapeutic uses of such stem and 
progenitor cells upon thawing. Such cells can be 
therapeutically valuable for hematopoietic reconstitution in 
patients with various diseases and disorders. In a preferred 
embodiment, neonatal cells that have been cryopreserved and 
thawed, can be used for autologous (self) hematopoietic 
reconstitution. 

The invention also relates to methods for collection and 
cryopreservation of the neonatal and fetal stem and 
progenitor cells of the invention, 

2, BACKGROUND OF THE INVENTION 

2,1, HEMATOPOIETIC STEM AND PROGENITOR CELLS 
The morphologically recognizable and functionally 
capable cells circulating in blood include erythrocytes, 
neutrophilic, eosinophilic, and basophilic granulocytes, B-, 
T-, nonB-, non T-lymphocytes , and platelets. These mature 
cells derive from and are replaced, on demand, by morphologi- 
cally recognizable dividing precursor cells for the 
respective lineages such as erythroblasts for the erythrocyte 
series, myeloblasts, promyelocytes and myelocytes for the 
granulocyte series, and megakaryocytes for the platelets. 
The precursor cells derive from more primitive cells that can 
simplistically be divided into two major subgroups: stem 
cells and progenitor cells (for review, see Broxmeyer, H.E., 
1983, "Colony Assays of Hematopoietic Progenitor Cells and 
Correlations to Clinical Situations," CRC Critical Reviews in 
Oncology/Hematology 1 (3) : 227-257) . The definitions of stem 
and progenitor cells are operational and depend on func- 
tional, rather than on morphological, criteria. Stem cells 
have extensive self-renewal or self-maintenance capacity 




(Lajtha, L.G., 1979, Differentiation 14:23), a necessity 
since absence or depletion of these cells could result in the 
complete depletion of one or more cell lineages, events that 
would lead within a short time to disease and death. Some of 
one stem cells differentiate upon need, but some stem cells 
or their daughter cells produce other stem cells to maintain 
the precious pool of these cells. Thus, in addition to 
maintaining their own kind, pluripotential stem cells are 
capable of differentiation into several sublines of 
progenitor cells with more limited self-renewal capacity or 
no self -renewal capacity. These progenitor cells ultimately 
give rise to the morphologically recognizable precursor 
cells. The progenitor cells are capable of proliferating and 
differentiating along one, or more than one, of the myeloid 
differentiation pathways (Lajtha, L.G. (Rapporteur), 1979, 
Blood Cells 5:447) . 

Stem and progenitor cells make up a very small percent- 
age of the nucleated cells in the bone marrow, spleen, and 
blood. About ten times fewer of these cells are present in 
the spleen relative to the bone marrow, with even less 
present in the adult blood. As an example, approximately one 
in one thousand nucleated bone marrow cells is a progenitor 
cell; stem cells occur at a lower frequency. These 
progenitor and stem cells have been detected and assayed for 
by placing dispersed suspensions of these cells into irradi- 
ated mice, and noting those cells that seeded to an organ 
such as the spleen and which found the environment conducive 
to proliferation and differentiation. These cells have also 
been quantified by immobilizing the cells outside of the body 
in culture plates (in vitro ) in a semi-solid support medium 
such as agar, methylcellulose, or plasma clot in the presence 
of culture medium and certain defined biomolecules or cell 
populations which produce and release these molecules. Under 
the appropriate growth conditions, the stem or progenitor 
cells will go through a catenated sequence of proliferation 



-8- 



and differentiation yielding mature end stage progeny, which 
thus allows the determination of the cell type giving rise to 
the colony. If the colony contains granulocytes, macropha- 
ges, erythrocytes, and megakaryocytes (the precursors to 
platelets, , then the cell giving rise to them would have been 
a pluripotential cell. To determine if these cells have 
self -renewal capacities, or sternness, and can thus produce 
more of their own kind, cells from these colonies can be 
replated in vivo or in vitro . Those colonies, which upon 
replating into secondary culture plates, give rise to more 
colonies containing cells of multilineages, would have 
contained cells with some degree of sternness. The stem cell 
and progenitor cell compartments are themselves heterogeneous 
with varying degrees of self-renewal or proliferative 
capacities. A model of the stem cell compartment has been 
proposed based on the functional capacities of the cell 
(Hellman, S., et al., 1983, J. Clin. Oncol. 1:227-284). 
Self -renewal would appear to be greater in those stem cells 
with the shortest history of cell division, and this self- 
renewal would become progressively more limited with 
subsequent division of the cells. 

A human hematopoietic colony-forming cell with the 
ability to generate progenitors for secondary colonies has 
been identified in human umbilical cord blood (Nakahata, T. 
and Ogawa, M. , 1982, J . Clin. Invest. 70:1324-1328). In 
addition, hematopoietic stem cells have been demonstrated in 
human umbilical cord blood, by colony formation, to occur at 
a much higher level than that found in the adult (Prindull, 
G., et al., 1978, Acta Paediatr. Scand. 67:413-416; Knudtzon, 
S., 1974, Blood 43 (3) : 357-361) . The presence of circulating 
hematopoietic progenitor cells in human fetal blood (Linch, 
D.C., et al., 1982, Blood 59 ( 5 ): 976-979 ) and in cord blood 
(Fauser, A. A. and Messner, H.A. , 1978, Blood 52 (6) : 1243-1248) 
has also been shown. Human fetal and neonatal blood has been 
reported to contain megakaryocyte and burst erythroblast 



35 




• - • 



-9- 



progenitors (Vainchenker, W. , et al., 1979, Blood Cells 
5:15-42), with increased numbers of erythroid progenitors in 
human cord blood or fetal liver relative to adult blood 
(Hassan, M.W., et al., 1979, Br. J. Haematol. 41:477-484; 
Tchernia, G., et ...1., 1981, J. Lab. Clin. Med. 97(3) :322- 
3 31) . Studies have suggested some differences between cord 
blood and bone marrow cells in the characteristics of CFU-GM 
(colony forming unit-granulocyte, macrophage) which express 
surface la antigens (Koizumi, S., et al - , 1982, Blood 
60(4) : 1046-1049) . 



suspensions comprising human stem and progenitor cells and 
methods for isolating such suspensions, and the use of the 
cell suspensions for hematopoietic reconstitution. 



2-2. RECONSTITUTION OF THE HEMATOPOIETIC SYSTEM 
Reconstitution of the hematopoietic system has been 
accomplished by bone marrow transplantation. Lorenz and 
coworkers showed that mice could be protected against lethal 
irradiation by intravenous infusion of bone marrow (Lorenz, 
E., et al., 1951, J. Natl. Cancer Inst. 12:197-201). Later 
research demonstrated that the protection resulted from 
colonization of recipient bone marrow by the infused cells 
(Lindsley, D.L., et al., 1955, Proc. Soc. Exp. Biol. Med. 
90:512-515; Nowell, P.C., et al., 1956, Cancer Res. 16:258- 
261; Mitchison, N.A. , 1956, Br. J. Exp. Pathol. 37:239-247; 
Thomas, E. D. , et al., 1957, N. Engl. J. Med. 257:491-496). 
Thus, stem and progenitor cells in donated bone marrow can 
multiply and replace the blood cells responsible for 
protective immunity, tissue repair, clotting, and other 
functions of the blood. In a successful bone marrow 
transplantation, the blood, bone marrow, spleen, thymus and 
other organs of immunity are repopulated with cells derived 
from the donor. 



10 



U.S. Patent No. 4,714,680 discloses cell 



15 



35 




-10- 



U.S. Patent No, 4,721,096 by Naughton et al. discloses a 
method of hematopoietic reconstitution which comprises 
obtaining and cryopreserving bone marrow, replicating the 
bone marrow cells in vitro , and then infusing the cells into 
a patient. 

Bone marrow has been used with increasing success to 
treat various fatal or crippling diseases, including certain 
types of anemias such as aplastic anemia (Thomas, E.D., et 
al., Feb. 5, 1972, The Lancet, pp. 284-289), Fanconi's anemia 
(Gluckman, E. , et al., 1980, Brit. J. Haematol. 45:557-564; 



Gluckman> E. , et al., 198 3, Brit. J. Haematol. 54:431-440; 
Gluckman, E. , et al., 1984, Seminars in Hematology : 21 
(l):20-26), immune deficiencies (Good, R.A- , et al., 1985, 
Cellular Immunol. 82:36-54), cancers such as lymphomas or 
leukemias (Cahn, J.Y., et al., 198 6, Brit. J. Haematol. 
63:457-470; Blume, K.J. and Forman, S.J., 1982, J. Cell. 
Physiol. Supp. 1:99-102; Cheever, M-A. , et al., 1982, N. 
Engl. J. Med. 307 (8) : 479-481) , carcinomas (Blijham, G. , et 
al., 1981, Eur. J. Cancer 17 (4) : 433-441) , various solid 
tumors (Ekert, H. , et al., 1982, Cancer 49:603-609; Spitzer, 
G., et al., 1980, Cancer 45:3075-3085), and genetic disorders 
of hematopoiesis. Bone marrow transplantation has also 
recently been applied to the treatment of inherited storage 
diseases (Hobbs, J.R., 1981, Lancet 2:735-739), thalassemia 
major (Thomas, E.D., et al. , 1982, Lancet 2:227-229), sickle 
cell disease (Johnson, F.J., et al., 1984, N. Engl. J. Med. 
311:780-783), and osteopetrosis (Coccia, P.F., et al., 1980, 
N. Engl. J. Med. 302:701-708) (for general discussions, see 
Storb, R. and Thomas, E.. D. , 1983, Immunol. Rev. 71:77-102; 
O'Reilly, R. , et al., 1984, Sem. Hematol . 21(3) : 188-221; 
1969, Bone-Marrow Conservation, Culture and Transplantation, 
Proceedings of a Panel, Moscow, July 22-26, 1968, 
International Atomic Energy Agency, Vienna; McGlave, P.B., et 
al., 1985, in Recent Advances in Haematology, Hoffbrand, 
A.V. , ed., Churchill Livingstone, London, pp. 171-197). 



10 



35 




-11- 



Present use of bone marrow transplantation is severely 
restricted, since it is extremely rare to have perfectly 
matched (genetically identical) donors, except in cases where 
an identical twin is available or where bone marrow cells of 
a patient in remission are stored in a viable frozen state. 
Even in such an autologous system, the danger due to 
undetectable contamination with malignant cells, and the 
necessity of having a patient healthy enough to undergo 
marrow procurement, present serious limitations. (For 
reviews of autologous bone marrow transplantation, see 



Herzig, R.H. , 1983, in Bone Marrow Transplantation, Weiner, 
R.S,, et al., eds. , The Committee On Technical Workshops, 
American Association of Blood Banks, Arlington, Virginia; 
Dicke, K.A. , et al. , 1984, Sem. Hematol. 21 (2 ): 109-122 ; 
Spitzer, G., et al., 1984, Cancer 54 (Sept. 15 Suppl . ) : 1216- 
122 5) . Except in such autologous cases, there is an 
inevitable genetic mismatch of some degree, which entails 
serious and sometimes lethal complications. These 
complications are two-fold. First, the patient is usually 
immunologically incapacited by drugs beforehand, in order to 
avoid immune rejection of the foreign bone marrow cells (host 
versus graft reaction) . Second, when and if the donated bone 
marrow cells become established, they can attack the patient 
(graft versus host disease) , who is recognized as foreign. 
Even with closely matched family donors, these complications 
of partial mismatching are the cause of substantial mortality 
and morbidity directly due to bone marrow transplantation 
from a genetically different individual. 

Peripheral blood has also been investigated as a source 
of stem cells for hematopoietic reconstitution (Nothdurtt, 
W. , et al., 1977, Scand. J. Haematol. 19:470-481; Sarpel, 
S.C., et al., 1979, Exp. Hematol. 71:113-120; Ragharachar, A. , 
et al., 1983, J. Cell. Biochem. Suppl. 7A:78; Juttner, C.A. , 
et al., 1985, Brit. J. Haematol. 61:739-745; Abrams, R.A., et 
al., 1983, J . Cell. Biochem. Suppl. 7A:53; Prummer, O., et 



10 



35 




10 



15 



20 



25 



30 



35 



-12- 



al-, 1985, Exp. Hematol . 13:891-898). In some studies, 
promising results have been obtained for patients with 
various leukemias (Reiffers, J., et al. , 1986, Exp. Hematol. 
14:312-315 (using cryopreserved cells); Goldman, J.M., et 
al., 1980, Br. J. Haematol. 45:223-231; Tilly, H., et al., 
July 19, 1986, The Lancet, pp. 154-155; see also To, L.B. and 
Juttner, C.A. , 1987, Brit. J. Haematol. 66: 285-288, and 
references cited therein); and with lymphoma (Korbling, M. , 
et al., 1986, Blood 67:529-532). It has been implied that 
the ability of autologous peripheral adult blood to 
reconstitute the hematopoietic system, seen in some cancer 
patients, is associated with the far greater numbers of 
circulating progenitor cells in the peripheral blood produced 
after cytoreduction due to intensive chemotherapy and/or 
irradiation (the rebound phenomenon) (To, L.B. and Juttner, 
C.A,, 1987, Annot. , Brit. J. Haematol. 66:285-288; see also 
1987, Brit. J. Haematol. 67:252-253, and references cited 
therein) . Other studies using peripheral blood have failed 
to effect reconstitution (Hershko, C, et al., 1979, The 
Lancet 1:945-947; Ochs, H.D., et al., 1981, Pediatr. Res. 
15(4 Part 2) :601) . 

Studies have also investigated the use of fetal liver 
cell transplantation (Cain, G.R., et al., 1986, 
Transplantation 41(l):32-25; Ochs, H.D., et al., 1981, 
Pediatr. Res. 15(4 part 2):601; Paige, C.J., et al., 1981, J. 
Exp. Med. 153:154-165; Touraine, J.L., 1980, Excerpta Med. 
514:277; Touraine, J.L., 1983, Birth Defects 19:139; see also 
Good, R.A., et al., 1983, Cellular Immunol. 82:44-45 and 
references cited therein) or neonatal spleen cell 
transplantation (Yunis, E. J. , et al., 1974, Proc. Natl. Acad. 
Sci. U.S.A. 72:4100) as stem cell sources for hematopoietic 
reconstitution. Cells of neonatal thymus have also been 
transplanted in immune reconstitution experiments (Vickery, 
A.C., et al., 1983, J. Parasitol. 69 (3) : 478-485 ; Hirokawa, 
K. , et al., 1982, Clin. Immunol. Immunopathol . 22:297-304), 



0 



10 



15 



20 



25 



30 



35 



-13- 



2.3. CRYQ PRESERVATION OF CELLS 
Freezing is destructive to most living cells. Upon 
cooling, as the external medium freezes, cells equilibrate by 
losing water, thus increasing intracellular t solute 
concentration. Below about 10-15*C, intracellular freezing 
will occur. Both intracellular freezing and solution effects 
are responsible for cell injury (Mazur, P., 1970, Science 
168:939-949). It has been proposed that freezing destruction 
from extracellular ice is essentially a plasma membrane 
injury resulting from osmotic dehydration of the cell 
(Meryman, H.T., et al . , 1977, Cryobiology 14:287-302). 

Cryoprotective agents and optimal cooling rates can 
protect against cell injury. Cryoprotection by solute 
addition is thought to occur by two potential mechanisms: 
colligatively , by penetration into the cell, reducing the 
amount of ice formed; or kinetically, by decreasing the rate 
of water flow out of the cell in response to a decreased 
vapor pressure of external ice (Meryman, H.T. , et al., 1977, 
Cryobiology 14:287-302). Different optimal cooling rates 
have been described for different cells. Various groups have 
looked at the effect of cooling velocity or cryopreservatives 
upon the survival or transplantation efficiency of frozen 
bone marrow cells or red blood cells (Lovelock, J.E. and 
Bishop, M.W.H., 1959, Nature 183:1394-1395; Ashwood-Smith , 
M.J., 1961, Nature 190:1204-1205; Rowe, A.W. and Rinfret, 
A. P., 1962, Blood 20:636; Rowe, A.W. and Fellig, J., 1962, 
Fed. Proc. 21:157; Rowe, A.W. , 1966, Cryobiology 3(1) : 12-18; 
Lewis, J. P., et al., 1967, Transfusion 7(l):17-32; Rapatz , 
G. , et al., 1968, Cryobiology 5(1): 18-25; Mazur, P., 1970, 
Science 168:939-949; Mazur, P., 1977, Cryobiology 14:251-272; 
Rowe, A.W. and Lenny, L.L., 1983, Cryobiology 20:717; Stiff, 
P.J., et al., 1983, Cryobiology 20:17-24; Gorin, N.C., 1986, 
Clinics in Haematology 15 (1) : 19-48) • 

The successful recovery of human bone marrow cells after 
long-term storage in liquid nitrogen has been described 



10 



15 



20 



25 



30 



35 



-14- 

(1983, American Type Culture Collection, Quarterly Newsletter 
3(4) :1). In addition, stem cells in bone marrow were shown 
capable of withstanding cryopreservation and thawing without 
significant cell death, as demonstrated by the ability to 
form equal numbers of mixed myeloid-erythroid colonies in 
vitro both before and after freezing (Fabian, I., et al., 
1982, Exp, Hematol. 10 ( 1) : 119-122) . The cryopreservation and 
thawing of human fetal liver cells (Zuckerman, A.J., et al., 
1968, J . Clin. Pathol. (London) 21 (1) : 109-110) , fetal 
myocardial cells (Robinson, D.M. and Simpson, J.F., 1971, In 
Vitro 6(5):378), neonatal rat heart cells (Alink, G.M. , et 
al., 1976, Cryobiology 13:295-304), and fetal rat pancreases 
(Kemp, J. A., et al., 1978, Transplantation 26 (4 ): 260-264) 
have also been reported. 

2.4. GENE THERAPY 
Gene therapy refers to the transfer and stable insertion 
of new genetic information into cells for the therapeutic 
treatment of diseases or disorders. The foreign gene is 
transferred into a cell that proliferates to spread the new 
gene throughout the cell population. Thus stem cells, or 
pluripotent progenitor cells, are usually the target of gene 
transfer, since they are proliferative cells that produce 
various progeny lineages which will potentially express the 
foreign gene. 

Most studies in gene therapy have focused on the use of 
hematopoietic stem cells. High efficiency gene transfer 
systems for hematopoietic progenitor cell transformation have 
been investigated for use (Morrow, J.F., 197 6, Ann. N.Y. 
Acad. Sci. 265:13; Salzar, W. , et al., 1981, in Organization 
and Expression of Globin Genes, A.R. Liss, Inc., New York, p. 
313; Bernstein, A., 1985, in Genetic Engineering: Principles 
-and Methods, Plenum Press, New York, p. 235; Dick, J.E., et 
al., 1986, Trends in Genetics 2:165). Reports on the 
development of viral vector systems indicate a higher 



-In- 



efficiency of transformation than DNA-mediated gene transfer 
procedures ( e.g. , CaP0 4 precipitation and DEAE dextran) and 
show the capability of integrating transferred genes stably 
in a wide variety of cell types. Recombinant retrovirus 
vectors have been widely used experimentally to transduce 
hematopoietic stem and progenitor cells* Genes that have 
been successfully expressed in mice after transfer by 
retrovirus vectors include human hypoxanthine phosphoribosyl 
transferase (Miller, A., et al., 1984, Science 255:630). 
Bacterial genes have also been transferred into mammalian 
cells, in the form of bacterial drug resistance gene 
transfers in experimental models. The transformation of 
hematopoietic progenitor cells to drug resistance by 
eukaryotic virus vectors, has been accomplished with recom- 
binant retrovirus-based vector systems (Hock, R.A. and 
Miller, A.D., 1986, Nature 320:275-277; Joyner, A, , et al-, 
1983, Nature 305:556-558; Williams, D.A- , et al-, 1984, 
Nature 310:476-480; Dick, J.E. , et al., 1985, Cell 42:71-79); 
Keller, G. , et al,, 1985, Nature 318:149-154; Eglitis, M. , et 
al., 1985, Science 230:1395-1398). Recently, adeno-associa- 
ted virus vectors have been used successfully to transduce 
mammalian cell lines to neomycin resistance (Hermonat, P.L. 
and Muzyczka, N., 1984, supra ; Tratschin, J.-D-, et al-, 
1985, Mol- Cell- Biol- 5:3251)- Other viral vector systems 
that have been investigated for use in gene transfer include 
papovaviruses and vaccinia viruses (see Cline, M-J., 1985, 
Pharmac- Ther. 29:69-92). 

Other methods of gene transfer include microinjection, 
electroporation, liposomes, chromosome transfer, and 
transfection techniques (Cline, M.J-, 1985, supra ) - Salser 
et al. used a calcium-precipitation transfection technique to 
transfer a methotrexate-resistant dihydrof olate reductase 
(DHFR) or the herpes simplex virus thymidine kinase gene, and 
a human globin gene into murine hematopoietic stem cells. In 
vivo expression of the DHFR and thymidine kinase genes in 



35 




-16- 



10 



15 



20 



25 



30 



35 



stem cell progeny was demonstrated (Salser, W. , et al. , 1981, 
in Organization and Expression of Globin Genes, Alan R. Liss, 
Inc., New York, pp. 313-334). 

Gene therapy has also been investigated in murine models 
with the goal of enzyme replacement therapy. Thus, normal 
stem cells from a donor mouse have been used to reconstitute 
the hematopoietic cell system of mice lacking beta- 
glucuronidase (Yatziv, S., et al., 1982, J. Lab. Clin. Med. 
90:792-797). Since a native gene was being supplied, no 
recombinant stem cells (or gene transfer techniques) were 
necessary. 

3. SUMMARY OF THE INVENTION 

The present invention is directed to hematopoietic stem 
and progenitor cells of neonatal or fetal blood, that are 
cryopreserved, and the therapeutic uses of such stem and 
progenitor cells upon thawing. In particular, the present 
invention relates to the therapeutic use of fetal or neonatal 
stem cells for hematopoietic (or immune) reconstitution. 
Hematopoietic reconstitution with the cells of the invention 
can be valuable in the treatment or prevention of various 
diseases and disorders such as anemias, malignancies, 
autoimmune disorders, and other immune dysfunctions and 
deficiencies • In another embodiment, fetal or neonatal 
hematopoietic stem and progenitor cells which contain a 
heterologous gene sequence can be used for hematopoietic 
reconstitution in gene therapy. 

In a preferred embodiment of the invention, neonatal or 
fetal blood cells that have been cryopreserved and thawed can 
be used for autologous (self) reconstitution. 

The invention also relates to methods of collection and 
cryopreservation of the neonatal and fetal stem and 
progenitor cells of the invention. 




-17- 

3.1. DEFINITIONS 
As used herein, the following abbreviations will have 
the meanings indicated: 

acid-citrate dextrose 

burst-forming unit-erythroid. An 
hematopoietic progenitor cell which is 
capable of producing a colony of 
erythroid progeny cells in semi-solid 
medium. 



ACD 

5 



BFU-E 



10 



15 



20 




BFU-E-1 = an early erythroid progenitor cell, 

capable of producing a colony of 
erythroid progeny cells in semi-solid 
medium upon stimulation by 
erythropoietin, hemin (optional) , and a 
burst-promoting factor . 

BFU-E-2 = an erythroid progenitor cell, of greater 

maturity than BFU-E-1, which is capable 
of producing a colony of erythroid 
progeny cells in semi-solid medium upon 
stimulation by erythropoietin and by 
hemin (optional) . 

CFU = colony-forming unit. A cell which is 

capable of producing a colony of progeny 
cells in semi-solid medium. 

CFU-GEMM = colony-forming unit-granulocyte , 

erythrocyte , monocyte/macrophage , 
megakaryocyte. A multipotent ial 
hematopoietic progenitor cell which is 
capable of producing a colony composed 

35 




-18- 

of granulocyte, erythrocyte, monocyte/ 
macrophage, and megakaryocyte progeny, 
in semi-solid medium. 




CFU-GM = colony-forming unit-granulocyte, 

5 

macrophage. An hematopoietic progenitor 
cell which is capable of producing a 
colony composed of granulocyte and 
macrophage progeny in semi-solid medium. 

10 

CFU-S = colony forming unxt-spleen. A 

multipotential stem cell with self- 
renewal capacity, which, upon 
inoculation into lethally-irradiated 
mice, is capable of producing a colony 
(nodule) on the spleen (s) . 



15 



CPD = citrate-phosphate-dextrose 

CSF = colony stimulating factor 

20 

DMSO = dimethyl sulfoxide 



25 



30 



DNase = deoxyribonuclease 

DPBS = phosphate buffered saline without 

magnesium or calcium 

FCS = fetal calf serum 

heterologous 

gene = a gene which is not present, or not 
functionally expressed, in the 
designated host cell. 



35 



• - • 



-19- 



IMDM 



= Iscove's Modified Dulbecco's Medium 



LD100/30 days = 



the minimum or near-minimal Lethal 
Dosage causing 100% mortality within a 
30-day post-irradiation period 



10 



PHALCM 



PWMSCM 



medium conditioned by 

phytohemagglutinin-stimulated leukocytes 
from patients with hemochromatosis 

pokeweed mitogen spleen cell conditioned 
medium 



15 



S-cell 



SLE 



= stem cell 



= systemic lupus erythematosus 



HTdr 



= tritiated thymidine 



20 



25 



30 



35 



TLI 



= total lymphoid irradiation 



4 . DESCRIPTION OF THE FIGURES 
Figure 1 presents data for neonatal blood volumes 
obtained in one series of collections from individual births. 
The volume (ml) of blood collected is shown along the X-axis, 
with infant weight (kg) along the Y-axis. Open circles 
represent births by Caesarian section; closed circles 
represent vaginal births. 

Figure 2 presents the data from neonatal blood volumes 
obtained in a second series of collections from individual 
births. The volume (ml) of blood collected is shown along 
the X-axis, with the infant weight (kg) along the Y-axis. 
Closed circles represent vaginal births, with collection by 
gravity drainage from the umbilical cord. Open circles 
represent births by Caesarian section, with collection by 
gravity drainage from the umbilical cord. Closed triangles 





-20- 



represent vaginal births, with collection from the delivered 
placenta. Open triangles represent births by Caesarian 
section, with collection from the delivered placenta. 

Figures 3A and 3B are diagrammatic representations of 
the composition of centrifuge tubes at different steps in a 
Ficoll-Hypaque density separation, as described in Section 
6.3.1, which can be employed to obtain low density cells that 
are enriched in hematopoietic stem and progenitor cells. The 
cord blood cell suspension is layered on Ficoll-Hypaque 
before centrif ugation (Fig. 3 A) . After centrifugation, the 
low density cells appear as a sharp band between the Ficoll- 
Hypaque and the phosphate-buffered saline (Fig. 3B) . 

Figure 4 is a diagrammatic representation of the 
apparatus described in Section 6.4, which can be used for the 
cryopreservation of neonatal and fetal hematopoietic stem and 
progenitor cells. The cryovials containing the cell 
suspensions are placed in a freezing rack which is in turn 
placed in a 4°C methanol bath. The methanol bath (in a metal 
or glass freezing dish) is in turn placed in a -80°C freezer. 
After the cells reach the frozen state, they are transferred 
to a long-term storage vessel containing liquid nitrogen. 

5. DETAILED DESCRIPTION OF THE INVENTION 
The present invention is directed to hematopoietic stem 
and progenitor cells of neonatal or fetal blood, that are 
cryopreserved, and the therapeutic uses of such stem and 
progenitor cells upon thawing. 

In particular, the present invention relates to the use 
of fetal or neonatal stem cells for hematopoietic reconstitu- 
tion. In a preferred embodiment of the invention, the fetal 
or neonatal stem cells can be used in autologous hematopoie- 
tic reconstitution, i.e. , in the reconstitution of the 
hematopoietic system of the same individual from which they 
were originally derived. In such an embodiment, the 
invention provides substantial advantages over the present 



• # 

-21- 



10 



15 



20 



25 



30 



use of bone marrow for hematopoietic reconstitution. Present 
use of bone marrow transplantation is severely restricted by 
the fact that there is virtually never a perfectly matched 
(genetically identical) donor, except in cases where an 
identical twin it> available or where bone marrow cells of, 
for example, a cancer patient in remission are stored in the 
viable frozen state in the hope that they will be free of 
malignant cells and healthy enough to be returned to the 
patient for treatment of any future relapse. Except in such 
cases, the inevitable genetic mismatch which results can 
entail the serious and sometimes lethal complications of host 
versus graft or graft versus host disease. In order to avoid 
host rejection of the foreign bone marrow cells (host versus 
graft reaction) , the patient must be immunologically 
incapacitated- Such immune incapacitation is itself a cause 
of serious complications. Furthermore, when and if the 
donated bone marrow cells become established, they can attack 
the patient (graft versus host disease) , who is recognized as 
foreign. Even with closely matched family donors, these 
complications of partial mismatching are the cause of 
substantial mortality and morbidity directly due to bone 
marrow transplantation from a genetically different 
individual . 

In an embodiment of the invention directed to the use of 
neonatal stem and progenitor cells for hematopoietic 
reconstitution, there are several main reasons for preferring 
the use of such neonatal cells to conventional bone marrow 
transplantation. First, no donor is required because the 
cells can be obtained from neonatal blood that would 
otherwise be discarded. Second, in a preferred autologous 
system, i.e. , involving use of 'self* neonatal cells, the 
complications arising in conventional bone marrow 1 
transplantation from the need for pretransplantation drug- 
induced or irradiation-induced immune incapacitation and from 
acute and chronic graf t-versus-host disease are all 




-22- 



eliminated because, in this embodiment, neonatal cells are 
returned to their original owner and are therefore totally 
compatible- For these reasons, present restrictions on the 
use of bone marrow transplantation arising from difficulties 
in finding even approximately matched donors, and from 
disease and mortality due to unavoidable genetic incompatibi- 
lity, do not apply to self-reconstitution with neonatal 
cells. Third, regarding the preferred autologous embodiment, 
the efficiency of genetically identical (self) cells in bone 
marrow transplantation in animals is numerically many times 
greater than that of cells from a genetically dissimilar 
donor (Balner, H. , 1977, Bone Marrow Transplantation and 
Other Treatment after Radiation Injury, Martinus Nijhoff 
Medical Division, The Hague) , thus far fewer self cells are 
required for successful reconstitution in the preferred 
autologous system. 

Furthermore, the prospects of success in bone marrow 
transplantation decline with age; although it is not clear 
whether the age of donor or patient is more important, it is 
proper to infer that younger (neonatal) cells are preferable 
for hematopoietic reconstitution. Such neonatal or fetal 
cells have not been subjected to the "environmental outrage" 
that adult cells have undergone. Also, as an example of 
novel medical applications which may be feasible with 
neonatal cells but not with conventional bone marrow 
transplantation, restoration with self cells taken at birth 
can be valuable in the treatment of disorders such as 
declining immune responsiveness and autoimmunity (immune 
reactions against one's own tissues) which occur in increas- 
ing frequency with age. 

Many of the relative disadvantages discussed supra of 
the use of bone marrow cells for hematopoietic 
reconstitution, also apply to the use of adult peripheral 
blood for such reconstitution, and thus, the use of neonatal 
cells for hematopoietic reconstitution according to the 



35 




-23- 



present invention provides distinct advantages over the 
employment of adult peripheral blood. It has been implied 
that the ability of autologous peripheral adult blood to 
reconstitute the hematopoietic system, seen in some cancer 
patients, is associated with the Jar greater numbers of 
circulating progenitor cells in the peripheral blood produced 
after cytoreduction due to intensive chemotherapy and/or 
irradiation (the rebound phenomenon) (To, L. B. and Juttner, 
C.A., 1987, Annot., Brit. *J. , Haematol, 66:285-288; see also 
1987, Brit. J. Haematol. 67:252-253, and references cited 
therein) . There are possible detrimental effects, known or 
unknown, of prior chemotherapy or irradiation, on the stem 
and progenitor cell populations found in these patients. 

There are additional reasons for preferring the use of 
neonatal cells for hematopoietic reconstitution as provided 
by the present invention. Neonatal blood is a preferred 
source of cells for hematopoietic reconstitution, since it is 
free from viral and microbial agents, known or unknown, 
latent or otherwise, that may be encountered in later life, 
other than those transmitted from the mother or during labor 
and delivery* In addition, in view of the extent to which 
the hematopoietic stem cell may possibly share with other 
cells the limitation in total number of cell divisions that 
it may undergo before senescence, it is proper to assume that 
the neonatal hematopoietic stem cell has a self-renewal and 
reconstituting capacity that is at least as great, and 
perhaps greater, than that of hematopoietic stem cells 
obtained at any later time in life. 

In adults, stem and progenitor cells are mostly confined 
to the bone marrow; very few circulate in the blood. In the 
newborn human or animal, however, stem and progenitor cells 
circulate in the blood in numbers similar to those found in 
adult bone marrow. Doubtless this reflects the great demands 
for blood formation of the growing infant. We calculate that 
the restorative capacity of neonatal blood contained in the 



35 






-24- 



human umbilical cord and placenta, which are customarily 
discarded at birth, equals or exceeds that of the average 
donation of an adult's bone marrow. The efficacy of human 
neonatal blood cells compared with adult bone marrow cells is 
gauged by laboratory assays for stem cell and progenitor 
cells. Progenitor cell assays imply that the reconstituting 
potential of cells from 50 ml of cord blood (readily obtain- 
able) is at least equivalent to the average number of 
progenitor cells from adult bone marrow that is used in 
autologous hematopoietic reconstitution (see Section 6.8, 
infra). 'S-cells', representing probably the earliest 
developmental form of the stem cell, are demonstrable in 
human (cord) blood (Nakahata, T. and Ogawa, M. , 1982, J. 
Clin, Invest. 70:1324-1328). Thus, the cells of neonatal 
blood can be judged an effective clinical substitute for 
adult bone marrow. 

In laboratory animals, the efficacy of neonatal cells 
can be tested directly. Accordingly we have shown that 
circulating neonatal cells, in numbers lower than are 
contained in the cord and placenta, will completely and 
permanently repopulate the entire blood-forming and immune 
systems of a lethally irradiated adult animal, promoting 
complete recovery and return to normal health (see Section 
6. 11, infra ) . 

The method of the invention may be divided into the 
following stages solely for the purpose of description: 
(a) isolation of fetal or neonatal hematopoietic stem and 
progenitor cells; (b) inspection and testing of fetal or 
neonatal blood; (c) enrichment for hematopoietic stem and 
progenitor cells; (d) cryopreservat ion ; (e) recovery of stem 
and progenitor cells from the frozen state; (f) examination 
of cells recovered for clinical therapy; and (g) therapeutic 
uses in reconstitution of the hematopoietic system. 

Since both fetal and neonatal hematopoietic cells are 
envisioned for use in the present invention, descriptions and 



35 




-25- 



10 



15 



20 



25 



30 



35 



embodiments of the invention herein described for neonatal 
cells are meant to apply equally to fetal cells, unless 
clearly otherwise indicated or apparent. 

5.1. ISOLATION OF FETAL OR NEONATAL 

HEMATOPOIETIC STEM AND PROGENITOR CELLS 

Fetal or neonatal blood are sources of the hematopoietic 
stem and progenitor cells of the present invention. 

Fetal blood can be obtained by any method known in the 
art. For example, fetal blood can be taken from the fetal 
circulation at the placental root with the use of a needle 
guided by ultrasound (Daffos, F. , et al., 1985, Am. J. 
Obstet. Gynecol. 153:655-660; Daffos, F. , et al., 1983, Am. 
J. Obstet. Gynecol. 146:985), by placentocentesis (Valenti, 
C, 1973, Am. J. Obstet. Gynecol. 115:851; Cao, A., et al., 
1982, J. Med. Genet. 19:81), by fetoscopy (Rodeck, C.H. , 
1984, in Prenatal Diagnosis, Rodeck, C.H. and Nicolaides, 
K.H., eds., Royal College of Obstetricians and Gynaecol- 
ogists, London), etc. 

In a preferred embodiment of the invention, neonatal 
hematopoietic stem and progenitor cells can be obtained from 
umbilical cord blood and/or placental blood. The use of cord 
or placental blood as a source of cells to repopulate the 
hematopoietic system provides numerous advantages. Cord 
blood can be obtained easily and without trauma to the donor. 
In contrast, at present, the collection of bone marrow cells 
for transplantation is a traumatic experience which is costly 
in terms of time and money spent for hospitalization. Cord 
blood cells can be used for autologous transplantation, when 
and if needed, and the usual hematological and immunological 
problems associated with the use of allogeneic cells, matched 
only partially at the major histocompatibility complex or 
matched fully at the major, but only partially at the minor 
complexes, are alleviated. 

Collections should be made under sterile conditions. 
Immediately upon collection, the neonatal or fetal blood 





-26- 



10 



15 



20 



25 



30 



should be mixed with an anticoagulent. Such an anti- 
coagulent can be any known in the art, including but not 
limited to CPD (citrate-phosphate-dextrose) , ACD (acid 
citrate-dextrose), Alsever's solution (Alsever, J.B. and 
Ainslie, R.B., 1941, N . Y . St, J. Med, 41:126)~, De Gowin'i- 
Solution (De Gowin, E.L., et al., 1940, J- Am. Med, Ass, 
114:850), Edglugate-Mg (Smith, W.W., et al., 1959, J • Thorac. 
Cardiovasc. Surg, 38:573), Rous-Turner Solution (Rous, P. 
and Turner, J.R,, 1916, J. Exp. Med. 23:219), other glucose 
mixtures, heparin, ethyl biscoumacetate, etc. (See Hurn, 
B.A.L., 1968, Storage of Blood, Academic Press, New York, pp. 
26-160) . In a preferred embodiment, ACD can be used. 

5.1.1. COLLECTION OF NEONATAL BLOOD 
The object of this aspect of the invention is to obtain 
a neonatal blood collection of adequate volume that is free 
of contamination. Since umbilical cord blood is a rich 
source of stem and progenitor cells (see Section 6.6, infra ; 
Nakahata, T. and Ogawa, M. , 1982, J. Clin. Invest. 70:1324- 
1328; Prindull, G. , et al , , 1978, Acta. Paediatr. Scand. 
67:413-416; Tchernia, G. , et al., 1981, J. Lab. Clin. Med. 
97 (3) : 322-331) , the preferred source for neonatal blood is 
the umbilical cord and placenta. The neonatal blood can 
preferably be obtained by direct drainage from the cord 
and/or by needle aspiration from the delivered placenta at 
the root and at distended veins. 



In a preferred embodiment, volumes of 50 ml or more of 
neonatal blood are obtained (see Section 6.1, infra). 

Practical experience indicates that volumes of 50 ml or 
more are easily collected without additional measures in 80% 
of term births, and that collections of more than 40 ml are 
obtainable more than 90% of the time. Lower volumes may also 
be acceptable, and indicated under some circumstances (see 




-27- 



10 



15 



20 



25 



30 



Sections 5.1.1.2.3-1 and 5.1.1.2.3.2, infra ) . 

The following information suggests that as little as 50 
ml of cord blood contains enough of the appropriate cells to 
repopulate the hematopoietic system of an adult, and it is 
possible that even less cord blood would have the same 
effect: 

1. In a small sampling of cases for autologous 
marrow transplantation (Spitzer, G. , et al M 1980, Blood 
55:317-323), rapid repopulation of hematopoiesis in patients 
with acute leukemia was associated with as few as 0.24 
million granulocyte-macrophage progenitor cells (CFU-GM) . 

2. In human cord blood, there are approximately 
50-200 CFU-GM per 100,000 low density cells and at least 5 
million low density cord blood cells per milliliter. Thus 50 
milliliters of cord blood would contain in the range of 0.1 
to greater than 0.5 million CFU-GM (see also Section 6.8, 
infra) . The upper value agrees closely with estimations from 
the number of CFU-GM in 12.5 to 19 day old fetal blood 
(Lynch, D.C., et al., 1982, Blood 59:976-979). 

3. Importantly, stem and progenitor cells in cord 
blood appear to have a greater proliferative capacity in 
culture dishes than those in adult bone marrow (Salahuddin, 
S.Z., et al., 1981, Blood 58:931-938; Cappellini, M.D., et 
al., 1984, Brit. J. Haematol. 57:61-70). 

Significant to the use of cord blood as a source of stem 
cells, is that the assay for S-cells has been adapted for the 
growth of human cord blood (Nakahata, T. and Ogawa, M. , 1982, 
J. Clin. Invest. 70:324-1328). All the known progenitor 
cells are present in cord blood in high numbers and this 
includes those progenitors for multilineages , granulocytes, 
macrophages, erythrocytes, mast cells, and basophils ( id . ; 
Fauser, A. A. and Messner, H.A. , 1978, Blood 52:1243-1248; 
Koizumi, S., et al., 1982, Blood 60:1046-1049; Prindull, G., 
et al., 1978, Acta Paediatr. Scand. 67:413-416). 



35 




10 



15 



20 



25 



30 



-28- 

Furthermore, hematopoietic stem and progenitor cells can 
potentially be multiplied in culture, before or after 
cryopreservation, (see Sections 5.1,3-2, 5.3.2, infra), thus 
expanding the number of stem cells available for therapy. 

5,1-1.2. PREFERRED ASPECTS 
The following subsections provide detailed descriptions 
of preferred particular embodiments of the invention, and are 
intended for descriptive purposes only, in no way limiting 
the scope of the invention. 

5.1.1-2.1. COLLECTION KIT 
In a preferred aspect, a collection kit, packaged in a 
sterile container, can be used. In one particular 
embodiment, the collection kit can consist of: 

(i) a wide-mouth, graduated, collection container, 
with anticoagulant, into which the cut end of the cord may be 
placed for collection by gravity drainage. A small funnel 
can be provided for use if needed. 

(ii) (optional) a plastic, flexible, sealed 
collection bag, similar to a donation bag, which has ports 
for injection of the collected blood, and contains anticoagu- 
lant. 

(iii) an identification label, which identifies 
the infant source of the sample and time of collection. 

For multiple births, separate collections, each 
performed with a separate kit, are preferred. 

Sterilization of the containers can occur by any 
technique known in the art, including but not limited to 
beta-irradiation, autoclaving of suitable materials in a 
steam sterilizer, etc. For example, in a preferred 
embodiment, sterilization by beta-irradiation can be carried 
out by exposure to 2.5 megarads from a tungsten source (see 
Section 6.1, infra) - 



35 



-29- 



The collection kit may be placed in the surgical field 
in advance of a delivery, to afford ready availability* 



5.1.1.2.2. 



VAGINAL DELIVERY OF THE TERM INFANT 



Vaginal delivery of the normal infant at term, spontane- 
ously, by forceps, or as a breech delivery, should allow an 
ample collection of cord blood. After clamping the cord, the 
volume of fetal blood remaining in the cord and attached 
placenta has been estimated at 4 5 ml/kg infant body weight, 
or approximately 145 ml for a 7 lb (3.2 kg) baby (Hellman, 
L.M. , et al., 1971, Williams Obstetrics, 14th Ed., Appleton- 
Century-Crofts, New York, p. 216) . 

Following delivery of the infant, by any method, with or 
without anesthesia, the infant is held in the plane of the 
vagina, and the cord is doubly cross-clamped and cut 
approximately three inches (7-8 cm) from the umbilicus. The 
infant is removed. 

Maintaining usual sterile precautions, the cord is then 
transected just above the crushed portion in the clamp, and 
the resulting flow of fetal blood from umbilical vessels is 
caught in the container provided- An adequate collection can 
usually be accomplished without milking the cord, and is 
complete in approximately two minutes, before placental 
separation has occurred. Care should be taken to avoid 
contamination by maternal blood, urine, or other fluids in 
the delivery field. Blood in the container is then 
transferred to the bag provided for transport to the storage 
facility or, alternatively, the original container, if 
equipped with a tight screw cap, can itself be sent to the 
storage facility without transfer of its contents. 

If, following infant delivery, events make collection at 
that time undesirable, collection can be done after delivery 
of the placenta (see Section 5.1.1.2.3.6, infra). If 
maternal infection is suspected, such a placental collection 
may be preferable. Collection can also be carried out by 



35 





-30- 



aspiration from the delivered placenta, in addition to 
gravity drainage. 

In a most preferred embodiment, immediate cord clamping 
after delivery is carried out, in order to achieve collection 
of the greatest possible volume of cord blood. Studies have 
shown that the relative distribution of blood between the 
infant and placental circuits gradually shifts to the 
infant's blood circuits with increasing delay in cord 
clamping after delivery (Yao, A.C., et al., October 25, 1969, 
Lancet: 871-873) . 



5.1.1.2.3. OTHER CIRCUMSTANCES OF BIRTH AND DELIVERY 

5.1.1.2. 3.1. PREMATURE BIRTH 

15 

The cord blood of premature infants may contain an even 

greater proportion of stem and progenitor cells than full- 
term cord blood. Consequently, smaller volumes of cord blood 
from premature infant delivery may give as good a yield of 
stem and progenitor cells. (The use of stem and progenitor 

20 

cell assays as described in Sections 5.4.2 and 6.6 can 
determine the yield) * Thus, in general, cord blood collec- 
tion should be carried out if premature infant survival is 
anticipated, even though the volume of blood collected may be 
less than usual. Collection procedures should be the same as 

25 

for term births. 



5.1.1.2.3.2. MULTIPLE BIRTHS 
Cord blood collections undertaken at the time of 
multiple births involve additional procedural considerations: 

30 

(i) Multiple births are often premature, and 
volumes of cord blood will be correspondingly smaller. 
Collections should be made nevertheless, so that the decision 
to preserve for storage can be made later. 



10 



15 



20 



25 



30 



35 



-31- 



(ii) When births of two or more infants occur, 
where use of the cord collection is envisioned for later 
sel f -reconst itut ion , it is essential that each cord 
collection be identified with the proper infant. In cases of 
..ioubtful zygosity, blood typing can be done on cord blood and. 
postnatal samples . 

(iii) The timing of twin cord blood collection 
can be at the discretion of the obstetrician (after delivery 
of one twin; or after delivery of both) . 

(iv) A careful description of the placental 
relationships should be made (single or double amnions; 
single, double or fused chorions) . 

5. -1. 1.2.3.3. CAESARIAN DELIVERY 
Cord blood collections at caesarean section can be 
carried out with the same kit, and with the same procedure, 
as vaginal delivery. The cut end of the cord is lowered to 
promote gravity drainage. 

At caesarean section, it is strongly preferred that the 
cord blood collection be made after delivery of the infant, 
and before placental separation. However, this may not be 
desirable in some instances, such as where there is brisk 
hemorrhage, the need to incise or separate an anteriorly 
implanted placenta, or preoccupation of personnel with other 
events in the operating field. Thus, in these and similar 
cases, the placenta can be removed, and cord blood collected 
from it later. 

5.1.1.2.3.4. COMPLICATED DELIVERY 
Complications of delivery arising from the condition of 
the mother or the infant, or both, may require the immediate 
and urgent attention of the obstetrician and his assistants. 
Under these circumstances, the delivered placenta can be 
placed to one side, and collection carried out as soon as 
feasible. 




-32- 



5.1.1.2.3.5. 



ABNORMAL PLACENTA 



For successful cord blood collection, it is preferred 
that the placenta be intact, or nearly so. Cases of marginal 
or partial separation can still offer an opportunity for 
collection, although it may have to be carried out after 
delivery of the placenta, if clinical circumstances indicate 
a need for prompt removal. Collections will be disfavored 
for use if a rupture of fetal circulation has occurred. 
Samples can be tested later for contamination by maternal 
blood (see Section 5.1.2, infra). Accurate description of 
the placental abnormality is preferred. 

5.1.1.2.3.6. COLLECTION FROM THE DELIVERED PLACENTA 
When rapid delivery of the placenta occurs or becomes 
necessary, and cord blood collection cannot be accomplished 
prior to placental separation, a sample of sufficient volume 
can still be obtained after delivery. The placenta and 
attached cord, still clamped, are placed to one side, but 
still within the sterile field. Collection is by the same 
technique described supra in section 5.1.1.2.2. It is 
preferred, however, that collection be completed within five 
minutes of delivery/ while maintaining sterile procedures. 

Cord blood collection prior to placental separation is 
preferred over collection from the delivered placenta for the 
following reasons: In a collection from delivered placenta, 
(i) collection volumes are generally less; (ii) some degree 
of clotting in the placental circulation may restrict 
recovery, and (iii) the likelihood of contamination, by 
maternal blood or other agents, is increased. Therefore, the 
determination of suitability of the sample collected from a 
delivered placenta is especially important. 

5.1.1.2.3.7. MEDICAL CONDITIONS OF THE MOTHER 
Given the general prohibition against maternal use of 
drugs which would adversely affect the fetus, it is unlikely 



35 




-33- 



that maternal therapy or medical status in the general sense 
would adversely affect stem cell retrieval from cord blood 
collection of a normal infant. In a preferred embodiment, 
however, specific information should be obtained in regard to 
drug abuse, vira. diseases capable of vertical transmission, 
and the influence of acute maternal illness at the time of 
delivery, since it is possible that these may affect stem 
cell retrieval from cord blood. 



Despite elaborate plans, delivery may occur inoppor- 
tunely, sometimes prematurely, and without the immediate 
services of a physician. Under these circumstances, the 
following procedures are preferred: (i) cord blood 
collection should be attempted with the standard kit, 
described supra ; (ii) the placenta, if delivered on an 
unsterile field, should simply be kept as clean as possible, 
left with the cord clamped, and collection attempted within 5 
minutes; (iii) the cord should be wiped with a cleansing 
agent ( e.g. Betadine) , and transected above the clamp, to 
make the collection; and (iv) circumstances of the delivery 
should be described with the specimen. 



In a preferred embodiment, the data listed in Table I, 
infra , are obtained at the time of collection in order to 
ensure the accurate identification and evaluation of the 
collected blood. 



5.1.1.2.3.8. 



UNPLANNED DELIVERY 



10 



5.1.1.2.4. 



RECORDATION OF DATA 



30 



35 




-34- 



TABLE I 

DATA TO BE RECORDED AT THE 
TIME OF NEONATAL BLOOD COLLECTION 

5 Date and time ot delivery 

Full name and address of mother 

Hospital identification 

Sex of infant 

Weight of infant 
10 Birth order (for multiple pregnancies) 

Gestational age 

Pregnancy complications 

Intrapartum complications 

Type of delivery 
15 Placental collection (amount of blood collected) 

Placental description and weight 

Condition of infant 



20 5.1.2. INSPECTION AND TESTING OF NEONATAL BLOOD 

In a preferred embodiment, the neonatal blood sample is 
inspected and tested to ensure its suitability. Appropriate 
inspections and tests include but are not limited to the 
procedures described infra. 

25 If the blood collection sample is to be shipped to a 

processing plant, the blood container and its contents should 
be inspected for defects such as inadequate closure and 
leakage. As an option, the collection kit may include a 
suitably positioned reusable maximum-minimum mercury 

3q thermometer to register the range of temperature change 

during shipment. Clots, opacity of the plasma and visible 
hemolysis are indications of bacterial contamination or other 
consequences of faulty handling. Time elapsed since 
collection can be noted. 

35 




n # - • 

-35- 



The following tests on the collected neonatal blood 
sample can be performed either routinely, or where clinically 
indicated: 

(i) Bacterial culture: To ensure the absence of 
microbial contamination, establis: led assays can be performed, 
such as routine hospital cultures for bacteria under aerobic 
and anaerobic conditions. 

(ii) Diagnostic screening for pathogenic microorganisms: 
To ensure the absence of specific pathogenic microorganisms, 
various diagnostic tests can be employed. Diagnostic 
screening for any of the numerous pathogens transmissible 
through blood can be done by standard procedures. As one 
example, the collected blood sample can be subjected to 
diagnostic screening for the presence of Human 

Immunodeficiency Virus (HIV) , the causative agent of Acquired 
Immune Deficiency Syndrome (AIDS) (Gallo et al. , 1984, 
Science 224:500-503; Barre-Sinoussi, F. , et al., 1983, 
Science 220:868; Levy, J. A. , et al., 1984, Science 225:840). 
Any of numerous assay systems can be used, based on the 
detection of virions, viral-encoded proteins, HIV-specific 
nucleic acids, antibodies to HIV proteins, etc. 

(iii) Confirmation of neonatal origin of the blood: 
Contamination with maternal blood, not necessarily a 
contraindication to storage and clinical utility, may be 
suspected from the obstetrical history. Presence of maternal 
cells, and of adult blood generally, can be revealed by 
various tests, including but not limited to I typing (Wiener, 
A.S., et al,, 1965, Am. J. Phys. Anthropol. 23(4): 389-396); 
analysis on a Coulter Channelyzer, which detects size 
differences between neonatal and maternal blood cells 
(Daffos, F., et al., 1985, Am. J. Obstet. Gynecol. 153:655- 
660) ; staining procedures for hemoglobin such as the 
Kleinhauer-Betke technique (Betke, K. , 1968, Bibl. 
Haematologica 29:1085) and others (Clayton, E.M., et al., 
1970, Obstetrics and Gynecology 35 ( 4 ) : 642-64 5 ) , which detect 



35 




-36- 



differences in the types of hemoglobin contained in red blood 
cells before birth versus in later life; etc. 

In a preferred embodiment, I typing can be done by 
established methods, such as agglutination with anti-i and 
anti-I antibodies. Erythrocytes of neonates are i strong, t 
weak; by 18 months of age, erythrocytes are I strong; i weak 
(Marsh, W.L., 1961, Brit- J. Haemat. 7:200). Thus, the 
degree of reaction with anti-i or anti-I antibodies is a 
measure of the proportion of neonatal blood and red cells in 
a mixture of neonatal and adult blood. The corresponding 



contamination with maternal stem and progenitor cells would 
be far less than the total maternal cell contamination since 
the stem and progenitor cells are rare in adult blood. 
(Scarcity of stem and progenitor cells in colony assays (see 
Sections 5.4.2 and 6.6, infra ) is another distinction between 
neonatal and adult blood. ) 



In a preferred embodiment of the invention, whole 
neonatal blood, as collected, can be cryogenically frozen, 
thus minimizing cell losses which can be incurred during cell 
processing protocols. However, cell separation procedures 
and expansion of stem and progenitor cells in in vitro 
cultures remain options. Such procedures may be useful, 
e.g. , in reducing the volume of sample to be frozen, and 
increasing cell count, respectively. The procedures 
described infra in Sections 5.1.3.1 and 5.1.3.2 should be 
carefully screened before use, in order to ensure that 
hematopoietic stem and progenitor cell loss in processing 
does not endanger the therapeutic efficacy of a collected 



10 



5.1.3. 



OPTIONAL PROCEDURES 



30 



blood sample in hematopoietic reconstitution. 



35 




-37- 



5.1.3.1. 



ENRICHMENT FOR HEMATOPOIETIC STEM AND 
PROGENITOR CELLS: CELL SEPARATION PROCEDURES 



After receiving cord blood or bone marrow samples in 

anticoagulant ( e.g. , ACD) , the cells can be subjected to 

physical and/or immunological cell separation procedures. 
5 - : 

Such procedures enrich for the hematopoietic stem and 

progenitor cells so that fewer total cells have to be stored 

and transplanted. However, if cell separation is desired, 

care should be taken to ensure sufficient recovery of the 

hematopoietic stem and progenitor cells. 

1 ^ Various procedures are known in the art and can be used 

to enrich for the stem and progenitor cells of the present 
invention. These include but are not limited to equilibrium 
density centrifugation, velocity sedimentation at unit 
gravity, immune rosetting and immune adherence, counter flow 

15 centrifugal elutriation, T lymphocyte depletion, and 

fluorescence-activated cell sorting, alone or in combination. 
Recently, procedures have been reported for the isolation of 
very highly enriched populations of stem/progenitor cells. 
Murine CFU-S have been purified by several groups using 

20 slightly different procedures (Visser, J.W.M., et al., 1984, 
J. Exp. Med. 59:1576; Nijhof, W. , et al., 1984, Exp. Cell 
Res. 155:583; Bauman, J.G.J. , et al., 1986, J. Cell. Physiol. 
128:133; Lord, B.I. and Spooncer, E., 1986, Lymphokine Res. 
5:59). Studies using human (Emerson, S.G., et al., 1985, J. 

25 Clin. Invest. 76:1286) or murine (Nicola, N.A. , et al., 1981, 
Blood 58:376) fetal liver cells have yielded highly enriched 
progenitor cells with up to 90% of them being colony forming 
cells for multi-, erythroid-, and granulocyte-macrophage 
lineages. CFU-E have also been very highly enriched (Nijhof, 

30 w., et al., 1983, J. Cell Biol. 96:386). Purification of 

adult mouse marrow CFU-GM with cloning efficiencies of up to 
99% in semi-solid medium has been accomplished by 
pretreatment of mice three days prior to sacrifice with 
cyclophosphamide, density separation of cells on Ficoll- 



35 




15 



-38- 

Hypaque, and counterflow centrifugal elutriation (Williams, 
D.E., et al., 1987, Exp. Hematol. 15:243). The resulting 
fraction of cells contained no detectable CFU-GEMM, BFU-E or 
CFU-MK, but up to 10% of the cells formed CFU-S measured at 
day 12, These procedures, or modifications thereof, can I a 

5 

used, and are within the scope of the present invention. 

Human stem and progenitor cells are present in the non- 
adherent, low density, T-lymphocyte-depleted fraction of bone 
marrow, spleen, and (adult and cord) blood cells. In a 

specific embodiment, low density (density less than 1.077 
10 3 

gm/cm ) cells can be separated by use of Fxcoll-Hypaque 
(Pharmacia Fine Chemicals, Piscataway, NJ) (see Section 
6.3.1, infra ) or Percol (Broxmeyer, H.E., 1982, J. Clin. 
Invest. 69:632-642). In this procedure, the mature cells of 
the granulocytic series, which are not needed for 
transplantation, are removed in the dense fraction which goes 
to the bottom of the tube. An adherence/nonadherence 
separation protocol can also be used for enrichment of 
hematopoietic stem and progenitors; protocols which can be 
used are described in Section 6.3.2, infra , and in Broxmeyer, 
H.E., et al., 1984, J. Clin. Invest. 73:939-953, which is 
incorporated by reference herein. 

If desired, autologous plasma can be removed for use in 
the freezing process. In particular, the blood or marrow 
samples can be allowed to settle at unit gravity in a test 
tube. The setting process can be hastened by addition of 
sterile-pyrogen-free Dextran Sulphate. After approximately 
15 minutes, the upper layer containing the nucleated cells in 
plasma can be removed and centrifuged ( e.g. , 200-400 X g) . 
The nucleated cells pellet to the bottom of the tube and the 
plasma is removed and stored in a tube at 4°C. The nucleated 
cells are washed, counted and, if desired, further separated 
( e.g, , by use of a density 'cut* procedure with Ficoll- 
Hypaque or Percol) . 

In order to enrich hematopoietic stem and progenitor 

35 



20 



25 



30 



i>7 



-39- 



cells, it is also possible to use cell separation procedures 
that entail immunological recognition of cells. Stem and 
progenitor cells can be isolated by positive or negative 
selection using antibodies that recognize antigenic 
determinants on the surface of cells. One means is to 
separate the cells by using monoclonal antibodies which 
recognize cell surface determinants on these cells, in 
conjunction with separation procedures such as fluorescence- 
activated cell sorting or panning (Broxmeyer, H.E., et al., 
1984, J* Clin. Invest. 73:939-953). At present, there are no 
known antigenic determinants that are absolutely specific for 
human hematopoietic stem and progenitor cells. However, 
these cells do contain antigenic determinants that are not 
present on all other cells, which can be used in antibody 
selection protocols for enrichment purposes; such antigens 
include but are not limited to those described infra . 

Within the human system, several antigens have been 
found on stem/progenitor cells. The first antigenic system 
studied intensively was that of the MHC class II antigens, 
especially HLA-DR. This has been found on CFU-GEMM, BFU-E, 
and CFU-GM (Lu, L. , et al., 198 3, Blood 61:250; Winchester, 
R.J., et al., 1977, Proc. Natl. Acad. Sci. U.S.A. 74:4012; 
Busch, F,W., et al., 1987, Blut 54:179). Several investigat- 
ors have suggested that HLA-DR are not found, or are present 
at a low density on cells earlier than CFU-GEMM (Moore, 
M.A.S., et al., 1980, Blood 55:682; Keating, A., et al., 
1984, Blood 64:1159) but others have not agreed ( e.g. , 
Falkenberg, J.H.F., et al., 1985, J. Exp. Med. 162:1359). 
This discrepancy may be due to the existence of specific 
subsets of early progenitors. In fact, the expression of 
HLA-DR is higher during the S-phase of the cell cycle of 
hematopoietic progenitor cells (Broxmeyer, H.E., 1982, J. 
Clin. Invest. 69:632; Cannistra, S.A., et al., 1985, Blood 
65:414). Day 14 CFU-GM express higher levels of HLA-DR than 
day 7 CFU-GM, and among day 7 CFU-GM, monocyte progenitors 



35 




-40- 



express more HLA-DR than do the granulocyte progenitors 
(Griffin, J.D., et al., 1985, Blood 66:788)- Expression of 
HLA-DR decreases and is lost during early myeloid precursor 
cell states and it has been suggested that HLA-DR antigens 
might play a role in myeloid development (Winchester , R.J . , 
et al., 1977, supra ) . 

Groups of antibodies have been used to distinguish 
different progenitors of the granulocyte-macrophage lineage 
(Ferrero, D. , et al., 1983, Proc. Natl, Acad. Sci. U.S.A. 
80:4114). Type 1 CFU-GM contribute all of the peripheral 
blood CFU-GM, as well as a small number of bone marrow CFU- 
GM. They express surface antigens recognized by S3-13 and 
S17-25 antibodies, but not by R1B19 and WGHS-29-1 antibodies. 
Type 2 CFU-GM are present only in the marrow and react with 
S3-13, R1B19, and WGHS-29-1. Culture of type 1 CFU-GM in 
liquid culture generates type 2 CFU-GM. These antibodies 
have also been used to characterize CFU-GM from patients with 
chronic myeloproliferative disorders (Robak, T. , et al., 
1985, Leukemia Res. 9:1023; Ferrero, D. , et al., 1986, Cancer 
Res. 46:975) . 

Other antigens on human stem/progenitor cells include 
those reactive with the MylO (Leary, A.G., et al., 1987, 
Blood 69:953; Strauss, L.C., et al . , 1986, Exp. Hematol. 
14:879), 3C5 (Katz, F.E., et al . , 1985, Leukemia Res. 9:191; 
Katz, F.E., et al., 1986, Leukemia Res. 10:961), RFB-1 
(Bodger, M.P., et al., 1983, Blood 61:1006), 12-8 (Andrews, 
R.G., et al., 1986, Blood 67:842), and L4F3 (Andrews, R.G. , 
et al., 1986, Blood 68:1030) antibodies. The antigen 
recognized by L4F3 is on CFU-GM, CFU-MK, BFU-E, and CFU-GEMM , 
but is apparently absent from cells which generate these 
progenitors in suspension culture ( id. ) . L4F3 reacts with 
most blast cells from patients with acute myelogenous 
leukemia, and treatment of cells from such patients with L4F3 
has allowed the growth of normal progenitor cells in vitro 
(Bernstein, I.D., et al., 1987, J. Clin. Invest. 79:1153). 



35 




-41- 



The antigen recognized by another antibody, Myll, is 
expressed on CFU-GM, but not on BFU-E or CFU-GEMM (Strauss, 
L.C., et al. , 1986, Exp. Hematol . 14:935). Receptors for 
various lectins are also expressed on stem/progenitors 
(Nicola, N.A., et al., 1980, J . Cell Physiol. 103:217; 
Reisner, Y. , et al., 1982, Blood 59:360; Reisner, Y. , et al., 
1978, Proc. Natl. Acad. Sci. U.S.A. 75:2933; Aizawa, S., and 
Tavassoli, M. , 1986, Int. J. Cell Cloning 4:464). 

Some success in enriching adult human bone marrow 
progenitor cells has been reported based on the use of 
monoclonal antibodies and cell sorting. In some studies, 
cells have been sorted only on positive versus negative 
populations (Katz, F.E., et al., 1986, Leukemia Res. 10:961). 
Recently, My 10 and HLA-DR antibodies were used in association 
with two color sorting to obtain highly enriched progenitor 
cell populations from human marrow (Lu, L. , et al., 1987, J. 
Immunol. 139(6) :1823-1829) . 

In specific embodiments, antibodies which are currently 
available and can be used in enrichment protocols include 
My-10, 3C5, or RFB-1. These antibodies can be used alone or 
in combination with procedures such as "panning" (Broxmeyer, 
H.E., et al., 1983, J. Clin. Invest. 73:939-953) or 
fluorescence activated cell-sorting (FACS) (Williams, D.E., 
et al., 1985, J. Immunol. 135:1004; Lu, L. , et al., 1986, 
Blood 68 (1) : 126-133) to isolate those cells containing 
surface determinants recognized by the monoclonal antibodies. 

In another embodiment, enrichment, if desired, can 
proceed by the use of monoclonal antibodies to major 
histocompatibility (MHC) class II antigens (especially HLA- 
DR) and to MylO (Lu, L. , et al., 1987, J. Immunol. 139(6): 
1823-1829. 

T lymphocyte depletion can also be used to enrich for 
hematopoietic stem or progenitor cells. In this procedure, T 
lymphocytes are selectively removed from the cell population 
by pretreating cells with a monoclonal antibody ( ies) , that 



35 






-42- 




10 



15 



20 



25 



30 



recognize a T cell antigen, plus complement:. Such a 
procedure has been described previously (Broxmeyer, H . E . , et 
al., 1984, J. Clin. Invest. 73:939-953). 

Another method that can be used is that of separating 
the stem and progenitor cells by means of selective agglu- 
tination using a lectin such as soybean (Reisner, Y. , et al., 
1980, Proc. Natl. Acad. Sci. U.S.A. 77:1164). This procedure 
can be a viable alternative for separation and enrichment of 
stem and progenitor cells without removal of possibly 
necessary accessory cells (Reisner, Y. , et al., 1983, Blood 
61(2) : 341-348; Reisner, Y. , et al . , 1982, Blood 59(2) :360- 
363) . 

Theoretically, only one early stem cell is needed for 
repopulation of the entire hematopoietic system. There is 
laboratory evidence that under ideal conditions and when the 
microenvironment nurturing the stem and progenitor cells in 
the recipient animal is not affected, a single stem cell can 
entirely repopulate the defective hematopoietic system of a 
mouse and rescue it from the lethal complications of anemia 
(Boggs, D.R. , et al . , 1982, J. Clin. Invest. 70:242-253). 
Doubtless, under clinical conditions in man it would 
generally require more than a single stem cell to rescue the 
hematopoietic system. Moreover, the presence of accessory or 
helper cells (non-stem/progenitor cells that influence the 
growth of stem/progenitor cells) , in addition to stem and 
progenitor cells, may be required (Spooncer, F. , et al. , 
1985, Nature (London) 316:62-64), especially if the 
microenvironment of the host is injured by treatments such as 
irradiation or chemotherapy. Thus, while there are ways to 
separate hematopoietic stem and progenitor cells from other 
cord blood cells (Leary, A.G. , et al., 1984, J. Clin. Invest. 
74:2193-2197) and these and other methods could be used to 
isolate and store pure or highly enriched preparations of 
these cells for transplantation, caution should be used in 
attempts at transplanting patients with purified preparations 




-43- 

of stem and progenitor cells, 

5.1,3,2 . IN VITRO CULTURES OF HEMATOPOIETIC 
STEM AND PROGENITOR CELLS 

An optional procedure (either before or after cryopres- . 
5 ervation) is to expand the hematopoietic stem and progenitor 
cells ijn vitro . However, care should be taken to ensure that 
growth in vitro does not result in the production of differ- 
entiated progeny cells at the expense of multipotent stem and 
progenitor cells which are therapeutically necessary for 

10 hematopoietic reconstitution. Various protocols have been 
described for the growth in vitro of cord blood or bone 
marrow cells, and it is envisioned that such procedures, or 
modifications thereof, may be employed (see Section 6*9 
infra; Smith, S, and Broxmeyer, H.E., 1986, Br, J. Haematol, 

15 63:29-34; Dexter, T.M, , et al., 1977, J • Cell. Physiol, 
91:335; Witlock, C.A, and Witte, O.N,, 1982, Prbc. Natl. 
Acad. Sci. U.S.A. 79:3608-3612). Various factors can also be 
tested for use in stimulation of proliferation in vitro , 
including but not limited to interleukin-3 (IL.-3) , 

20 granulocyte-macrophage (GM) -colony stimulating factor (CSF) , 
IL-1 (hemopoietin-1) , IL-4 (B cell growth factor) , IL-6, 
alone or in combination. 

5.2. CRYOPRESERVATION 
25 The freezing of cells is ordinarily destructive. On 

cooling, water within the cell freezes. Injury then occurs 
by osmotic effects on the cell membrane, cell dehydration, 
solute concentration, and ice crystal formation. As ice 
forms outside the cell, available water is removed from 
30 solution and withdrawn from the cell, causing osmotic 

dehydration and raised solute concentration which eventually 
destroy the cell. (For a discussion, see Mazur, P., .1977, 
Cryobiology 14:251-272.) 

These injurious effects can be circumvented by (a) use 

35 




-44- 



of a cryoprotective agent, (b) control of the freezing rate, 
and (c) storage at a temperature sufficiently low to minimize 
degradative reactions . 

Cryoprotective agents which can be used include but are 
not limited to dimethyl sulfoxide (DMSO) (Lovelock, J.E. and 
Bishop, M.W.H., 1959, Nature 183:1394-1395; Ashwood-Smith, 
M.J., 1961, Nature 190:1204-1205), glycerol, 
polyvinylpyrrolidine (Rinfret, A. P., 1960, Ann. N. Y. Acad. 
Sci. 85:576), polyethylene glycol (Sloviter, H.A. and Ravdin, 
R.G., 1962, Nature 196:548), albumin, dextran, sucrose, 
ethylene glycol, i-erythritol , D-ribitol, D-mannitol (Rowe, 
A.W., et al., 1962, Fed. Proc. 21:157), D-sorbitol, 
i-inositol, D-lactose, choline chloride (Bender, M.A. , et 
al., 1960, J. Appl. Physiol. 15:520), amino acids (Phan The 
Tran and Bender, M.A. , 1960, Exp. Cell Res. 20:651), 
methanol, acetamide, glycerol monoacetate (Lovelock, J.E., 
1954, Biochem. J. 56:265), and inorganic salts (Phan The Tran 
and Bender, M.A. , 1960, Proc. Soc. Exp. Biol. Med. 104:388; 
Phan The Tran and Bender, M.A. , 1961, in Radiobiology , 
Proceedings of the Third Australian Conference on 
Radiobiology, Ilbery, P.L.T., ed. , Butterworth, London, p. 
59) . In a preferred embodiment, DMSO is used, a liquid which 
is nontoxic to cells in low concentration. Being a small 
molecule, DMSO freely permeates the cell and protects 
intracellular organelles by combining with water to modify 
its freezability and prevent damage from ice formation. 
Addition of plasma ( e.g. , to a concentration of 20-25%) can 
augment the protective effect of DMSO. After addition of 
DMSO, cells should be kept at 0*C until freezing, since DMSO 
concentrations of about 1% are toxic at temperatures above 
4 °C. 

A controlled slow cooling rate is critical. Different 
cryoprotective agents (Rapatz, G., et al., 1968, Cryobiology 
5(l):18-25) and different cell types have different optimal 
cooling rates ( see e.g. , Rowe, A.W. and Rinfret, A. P., 1962, 



35 






-45- 



10 



15 



20 



25 



30 



Blood 20:636; Rove, A.W., 1966, Cryobiology 3(1): 12-18; 
Lewis, J, P., et al., 1967, Transfusion 7(1): 17-32; and Mazur, 
P., 1970, Science 168:939-949 for effects of cooling velocity 
on survival of marrow-stem cells and on their transplantation 
potential) . Th<_ heat of fusion phase where water turns to 
ice should be minimal. The cooling procedure can be carried 
out by use of, e.g. , a programmable freezing device or a 
methanol bath procedure. 

Programmable freezing apparatuses allow determination of 
optimal cooling rates and facilitate standard reproducible 
cooling. Programmable control led-rate freezers such as 
Cryomed or Planar permit tuning of the freezing regimen to 
the desired cooling rate curve. For example, for marrow 
cells in 10% DMSO and 20% plasma, the optimal rate is 1 to 
3°C/minute from 0'C to -80°C. In a preferred embodiment, 
this cooling rate can be used for the neonatal cells of the 
invention. The container holding the cells must be stable at 
cryogenic temperatures and allow for rapid heat transfer for 
effective control of both freezing and thawing. Sealed 
plastic vials ( e.g. , Nunc, Wheaton cryules) or glass ampules 
can be used for multiple small amounts (1-2 ml) , while larger 
volumes (100-200 ml) can be frozen in polyolefin bags ( e.g. , 
Delmed) held between metal plates for better heat transfer 
during cooling. (Bags of bone marrow cells have been 
successfully frozen by placing them in -80 °C freezers which, 
fortuitously, gives a cooling rate of approximately 
3 *C/minute) . 

In an alternative embodiment, the methanol bath method 
of cooling can be used. The methanol bath method is well- 
suited to routine cryopreservation of multiple small items on 
a large scale. The method does not require manual control of 
the freezing rate nor a recorder to monitor the rate. In a 
preferred aspect, DMSO-treated cells are precooled on ice and 
transferred to a tray containing chilled methanol which is 
placed, in turn, in a mechanical refrigerator ( e.g. , Harris 




-46- 



or Revco) at -80 °C Thermocouple measurements of the 
methanol bath and the samples indicate the desired cooling 
rate of 1 to 3°C/minute. After at least two hours, the 
specimens have... reached a temperature of -80°C and can be 
placed directly into liquid nitrogen (-196*C) for permanent 
storage* 

After thorough freezing, cells can be rapidly trans- 
ferred to a long-term cryogenic storage vessel . In a 
preferred embodiment, samples can be cryogenically stored in 
liquid nitrogen (-196*C) or its vapor (-165°C). Such storage 
is greatly facilitated by the availability of highly 
efficient liquid nitrogen refrigerators, which resemble large 
Thermos containers with an extremely low vacuum and internal 
super insulation, such that heat leakage and nitrogen losses 
are kept to an absolute minimum. 

In a particular embodiment, the cryopreservation 
procedure described in Section 6*4 infra is envisioned for 
use. The sterilized storage cryules preferably have their 
caps threaded inside, allowing easy handling without 
contamination. Suitable racking systems are commercially 
available and can be used for cataloguing, storage, and 
retrieval of individual specimens. 

Considerations and procedures for the manipulation, 
cryopreservation, and long-term storage of hematopoietic stem 
cells, particularly from bone marrow or peripheral blood, is 
largely applicable to the neonatal and fetal stem cells of 
the invention. Such a discussion can be found, for example, 
in the following references, incorporated by reference 
herein: Gorin, N.C., 1986, Clinics In Haematology 15(1) :19- 
48; Bone-Marrow Conservation, Culture and Transplantation, 
Proceedings of a Panel, Moscow, July 22-26, 1968, 
International Atomic Energy Agency, Vienna, pp. 107-186. 

Other methods of cryopreservation of viable cells, or 
modifications thereof, are available and envisioned for use 
( e.g. , cold metal-mirror techniques; Livesey, S.A. and 



35 





-47- 

Linner, J.G., 1987, Nature 327:255; Linner, J.G. , et al., 
1986, J, Histochem. Cytochem. 34 (9) : 1123-1135 ; see. also U.S. 
Patent No. 4,199,02 2 by Senkan et al., U.S. Patent No. 
3,753,357 by Schwartz, U.S. Patent No. 4,559,298 by Fahy; and 
Section 2.3, supra ) . 

5.3. RECOVERING STEM AND PROGENITOR 
CELLS FROM THE FROZEN STATE 

5.3.1. THAWING 

Frozen cells are preferably thawed quickly ( e.g. , in a 

10 water bath maintained at 37-41 °C) and chilled immediately 

upon thawing. In particular, the vial containing the frozen 
cells can be immersed up to its neck in a warm water bath; 
gentle rotation will ensure mixing of the cell suspension as 
it thaws and increase heat transfer from the warm water to 

15 the internal ice mass. As soon as the ice has completely 
melted, the vial can be immediately placed in ice (see 
Section 6.5, infra ) . 

5.3.2. OPTIONAL PROCEDURES 

20 in a preferred embodiment of the invention, the neonatal 

blood sample as thawed can be infused for hematopoietic 
reconstitution. Thus, it is envisioned that whole neonatal 
blood, cryopreserved and thawed, can be infused for therapy. 
However, several procedures, relating to processing of the 

25 thawed cells are available, and can be employed if deemed 
desirable. Such procedures are discussed infra . 

It may be desirable to treat the cells in order to 
prevent cellular clumping upon thawing. To prevent clumping, 
various procedures can be used, including but not limited to, 

30 the addition before and/or after freezing of DNase (Spitzer, 
G., et al., 1980, Cancer 45:3075-3085), low molecular weight 
dextran and citrate, hydroxyethyl starch (Stiff, P. J. , et 
al., 1983, Cryobiology 20:17-24), etc. 

The cryoprotective agent, if toxic in humans, should be 

35 




10 



15 



20 



25 



30 



-48- 

removed prior to therapeutic use of the thawed neonatal stem 
and progenitor cells. In an embodiment employing DMSO as the 
cryopreservative, it is preferable to omit this step in order 
to avoid cell loss, since DMSO has no serious toxicity. 
However, where removal of the cryoprotec :ive agent is 
desired, the removal is preferably accomplished upon thawing. 

One way in which to remove the cry ©protective agent is 
by dilution to an insignificant concentration. This can be 
accomplished by addition of medium, followed by, if 
necessary, one or more cycles of centrifugation to pellet 
cells, removal of the supernatant, and resuspension of the 
cells. For example, intracellular DMSO in the thawed cells 
can be reduced to a level (less than 1%) that will not 
adversely affect the recovered cells. This is preferably 
done slowly to minimize potentially damaging osmotic 
gradients that occur during DMSO removal (see Section 6,5, 
infra . ) 

After removal of the cryoprotective agent, cell count 
( e.g. , by use of a hemocytometer) and viability testing 
( e.g. , by trypan blue exclusion; Kuchler, R.J. 1977, 
Biochemical Methods in Cell Culture and Virology, Dowden, 
Hutchinson & Ross, Stroudsburg, Pa., pp. 18-19; 1964, Methods 
in Medical Research, Eisen, H.N., et al., eds., Vol. 10, Year 
Book Medical Publishers, Inc., Chicago, pp. 39-47) can be 
done to confirm cell survival. 

Other procedures which can be used, relating to 
processing of the thawed cells, include enrichment for 
hematopoietic stem and progenitor cells (see Section 5.1.3.1, 
supra ) and expansion by in vitro culture (see Section 
5.1.3.2, supra ) . However, in a preferred embodiment, these 
steps can be omitted in order to minimize cell loss. 



5.4. EXAMINATION OF CELLS RECOVERED 
FOR CLINICAL THERAPY 

In a preferred, but not required, aspect of the 

35 



41 



10 



15 



20 



25 



-49- 



invention, thawed cells are tested by standard assays of 
viability ( e.g. , trypan blue exclusion) and of microbial 
sterility (see Section 5.1.2, supra ) , and tested to confirm 
and/or determine their identity relative to the patient, and 
for hematopoietic function. 

5.4.1. IDENTITY TESTING 

Methods for identity testing which can be used include 
but are not limited to HLA (the major histocompatibility 
complex in man) typing (Bodmer, W. , 1973, in Manual of Tissue 
Typing Techniques, Ray, J.G., et al., eds., DHEW Publication 
No. (NIH) 74-545, pp* 24-27), and DNA fingerprinting, which 
can be used to establish the genetic identity of the cells. 
DNA fingerprinting (Jeffreys, A.J., et al., 1985, Nature 
314:67-73) exploits the extensive restriction fragment length 
polymorphism associated with hypervariable minisatellite 
regions of human DNA, to enable identification of the origin 
of a DNA sample, specific to each individual (Jeffreys, A.J. , 
et al., 1985, Nature 316:76; Gill, P., et al., 1985, Nature 
318:577; Vassart, G. , et al., 1987, Science 235:683), and is 
thus preferred for use. 

In a specific embodiment of the invention in which the 
cells recovered for therapy are to be used in an autologous 
system, the cells should match exactly the recipient patient 
from whom they originally came. 

5.4.2. ASSAYS FOR STEM AND PROGENITOR CELLS 



Any of numerous assays for hematopoietic stem or 
progenitor cells may be used (see Section 2.1). Examples of 
specific assays are described in Section 6.6 and subsections, 
30 infra . Modifications of the assays therein described are 

also envisioned for use. For example, various factors, alone 
or in combination, can be tested for stimulation of colony 
formation upon inclusion in the culture mixture (se£ 
Broxmeyer, H.E., 1986, Int. J. Cell Cloning 4:378-405; Lu, L. 

35 





-50- 



and Broxmeyer, H.E., 1983, Exp. Hematol. 11 (8) : 721-729 ; Lu, 
L. and Broxmeyer, H.E., 1985, Exp, Hematol. 13:989-993); such 
factors include but are not limited to oxygen tension, E-type 
prostaglandins, interleukin-3 (IL-3) , granulocyte-macrophage 
(GM) -colony stimulating factor (CSF) , granulocyte (G) -CSF-, 
macrophage (M)-CSF (CSF-1) , erythropoietin, IL-1, IL-4 (B 
cell growth factor) , hemin (ferric chloride protoporphyrin 
IX) , and media conditioned by various cell types. Culture 
assay methods may thus be changed to employ more efficient 
conditions for colony growth. In addition to in vitro colony 
forming assays, a stem cell assay for CFU-S (colony forming 
unit-spleen) can be done. In this assay, cells considered to 
be multipotential stem cells with self -renewal capacity can 
be measured by counting the number of colonies (nodules) on 
the spleen (s) of lethally-irradiated mice that have been 
inoculated with a composition containing the cells. 

In a particular embodiment, low density Ficoll-Hypaque- 

3 

separated cells (density less than 1,077 gm/cm ), which 

include the stem and progenitor cells, are plated, usually 
5 

0.5-2,0 x 10 per plate, for recognition of S (stem) cells, 
and progenitor cells of the CFU-GEMM (multipotent ) and CFU-GM 
and BFU-E (more differentiated) categories. 



The neonatal hematopoietic stem and progenitor cells of 
the present invention can be used therapeutically for 
hematopoietic reconstitution, with either syngeneic or 
allogeneic hosts. The neonatal cells can be introduced into 
a patient for repopulation of the blood and other 
hematopoietic organs in the treatment or prevention of 
various diseases or disorders, as described infra in Section 
5.6. Introduction of the neonatal cells can occur by any 
method known in the art, with systemic infusion of cells 
being the preferred route. 



5.5 



HEMATOPOIETIC RECONSTITUTION 



35 





-51- 



In a preferred embodiment of the invention, the neonatal 
cells are autologous (self) cells, i.e. , the cells were 
originally derived from the host recipient. Such an 
embodiment avoids the immunosuppressive regimens ( e.g. , 
irradiation, chemotherapy) which are often necessary in 
allogeneic transplants in order to avoid debilitating graft 
versus host or host versus graft disease. 



10 



15 



20 



25 



30 



5.6. THERAPEUTIC USES 

Reconstitution of the hematopoietic system (or immune 
system) with the neonatal stem and progenitor cells of the 
present invention can be therapeutically valuable for a large 
number of diseases and disorders. 

In a preferred embodiment involving the use of 
autologous neonatal cells, the infusion of previously 
cryopreserved neonatal hematopoietic stem and progenitor 
cells for hematopoietic reconstitution at any time after 
birth can not only be applied in the treatment of diseases 
which are presently known to be curable by allogeneic bone 
marrow transplantation, but also offers therapeutic potential 
for a number of additional diseases which presently are not 
considered likely to benefit from allogeneic marrow 
transplantation. This is due to the fact that allogeneic 
marrow transplantation (except for the few patients who are 
already immunologically incompetent) requires 
pretransplantation conditioning of the recipient with 
intensive cytoreduction with irradiation or chemotherapy for 
the purpose of eliminating the host (recipient) immune system 
in order to allow the transplanted marrow cells to engraft. 
This pretransplantation cytoreduction in combination with 
allogeneic HIA-ident ical marrow transplantation can result in 
a number of serious transplantation-induced complications 
such as life-threatening infections, long-lasting 
immunodeficiencies , and frequently, graf t-versus-host 
disease . 



35 



£2 




-52- 



Disorders that can be treated by infusion of stem cells 
include but are not limited to five broad categories. First 
are diseases resulting from a failure or dysfunction of 
normal blood cell production and maturation ( i. e. , aplastic 
anemia and hypoprol iterative stem cell disorders). The 
second group are neoplastic, malignant diseases in the 
hematopoietic organs ( e.g. , leukemia and lymphomas) . The 
third group of disorders comprises those of patients with a 
broad spectrum of malignant solid tumors of non-hematopoietic 
origin. Stem cell infusion in these patients serves as a 
bone marrow rescue procedure, which is provided to a patient 
following otherwise lethal chemotherapy or irradiation of the 
malignant tumor. The fourth group of diseases consists of 
autoimmune conditions, where the stem cells serve as a source 
of replacement of an abnormal immune system. The fifth group 
of diseases comprises a number of genetic disorders which can 
be corrected by infusion of hematopoietic stem cells, 
preferably syngeneic, which prior to transplantation have 
undergone gene therapy. Particular diseases and disorders 
which can be treated by hematopoietic reconstitution with 
neonatal stem and progenitor cells include but are not 
limited to those listed in Table II, and described infra . 



25 



30 



35 



53 



-53- 



10 




TABLE II 

DISEASES OR DISORDERS WHICH CAN BE 
TREATED BY HEMATOPOIETIC RECONSTITUTION 
WITH NEONATAL STEM AND PROGENITOR CELLS 



I. Diseases resulting from a failure or dysfunction of 
normal blood cell production and maturation 

hyperprolif erative stem cell disorders 
aplastic anemia 
pancytopenia 
agranulocytos is 
thrombocytopenia 
red cell aplasia 
15 Blackf an-Diamond syndrome 

due to drugs, radiation, or infection 
idiopathic 

II. Hematopoietic malignancies 

20 acute lymphoblastic (lymphocytic) leukemia 

chronic lymphocytic leukemia 
acute myelogenous leukemia 
chronic myelogenous leukemia 
acute malignant myelosclerosis 
multiple myeloma 
polycythemia vera 
agnogenic myelometaplasia 
Waldenstrom ' s macroglobul inemia 
Hodgk i n ' s 1 ymphoma 
30 non-Hodgkins's lymphoma 

III. Immunosuppression in patients with malignant, solid 
tumors 

ma 1 ignan t me 1 anoma 

35 




-54- 

carcinoma of the stomach 

ovarian carcinoma 

breast carcinoma 

small cell lung carcinoma 

retinoblastoma 

testicular carcinoma 

glioblastoma 

rhabdomyosarcoma 

neuroblastoma 

Ewing's sarcoma 

lymphoma 

IV • Autoimmune diseases 

rheumatoid arthritis 
diabetes type I 
chronic hepatitis 
multiple sclerosis 
systemic lupus erythematosus 

Genetic (congenital) disorders 
anemias 

familial aplastic 
Fanconi ' s syndrome 
Bloom's syndrome 
pure red cell aplasia (PRCA) 
dyskeratosis congenita 
Blackf an-Diamond syndrome 

congenital dyserythropoietic syndromes I-IV 
Chwachmann-Diamond syndrome 
dihydrofolate reductase deficiencies 
formamino transferase deficiency 
Lesch-Nyhan syndrome 
congenital spherocytosis 
congenital elliptocytosis 
congenital stomatocytosis 



V, 

20 



25 



30 



-55- 



congenital Rh null disease 
paroxysmal nocturnal hemoglobinuria 
G6PD (glucose-6-phosphate dehydrogenase) 

variants 1,2,3 
v pyruvate kinase deficiency 

congenital erythropoietin sensitivity 

deficiency 
sickle cell disease and trait 
thalassemia alpha, beta, gamma 
met-hemoglobinemia 

10 

congenital disorders of immunity 

severe combined immunodeficiency disease 
(SCID) 

bare lymphocyte syndrome 

ionophore- r espons ive combined 

1*5 ... 

immunode f ic lency 

combined immunodeficiency 

with a capping abnormality 

nucleoside phosphorylase deficiency 

granulocyte actin deficiency 

20 . . 

infantile agranulocytosis 

Gaucher ' s disease 

adenosine deaminase deficiency 

Kostmann ' s syndrome 

reticular dysgenesis 

25 

congenital leukocyte dysfunction syndromes 



VI . Others 

osteopetrosis 
myelosclerosis 
acquired hemolytic anemias 
acquired immunodeficiencies 
infectious disorders causing primary or 
secondary immunodeficiencies 
bacterial infections ( e.g. , Brucellosis, 



6h 



10 



15 



20 



25 



30 



• 



-56- 



Listerosis, tuberculosis, leprosy) 

parasitic infections ( e.g. , malaria, 
Leishmaniasis) 

.fungal infections 
disorders involving disproportions in 

lymphoid cell sets and impaired 

immune functions due to aging 
phagocyte disorders 

Kostmann' s agranulocytosis 

chronic granulomatous disease 

Chediak-Higachi syndrome 

neutrophil act in deficiency 

neutrophil membrane GP-180 deficiency 
metabolic storage diseases 

mucopolysaccharidoses 

mucolipidoses 
miscellaneous disorders involving 

immune mechanisms 
Wiskott-Aldrich Syndrome 
alpha 1-antitrypsin deficiency 



35 



5.6.1. DISEASES RESULTING FROM A FAILURE 
OR DYSFUNCTION OF NORMAL BLOOD 
CELL PRODUCTION AND MATURATION 

In this embodiment of the invention, reconstitution of 
the hematopoietic system with neonatal stem and progenitor 
cells can be used to treat diseases resulting from a failure 
or dysfunction of normal blood cell production and matura- 
tion, i.e. , aplastic anemia and hypoprolif erative stem cell 
disorders. These disorders entail failure of stem cells in 
bone marrow to provide normal numbers of functional blood 
cells. The aplastic anemias result from the failure of stem 
cells to give rise to the intermediate and mature forms of 
red cells, white cells, and platelets. Red cell production 
is usually most seriously affected, but a marked decrease in 




-57- 



production of other mature blood cell elements is also seen. 
The large majority of these anemias are acquired during adult 
life, and do not have any apparent genetic predisposition. 
About half of these acquired anemias arise in the absence of 
any obvious causative factor such as exposure to poisons, 
drugs or disease processes that impair stem cell function; 
these are termed idiopathic aplastic anemias. The remaining 
cases are associated with exposure to an extremely diverse 
array of chemicals and drugs and can also occur as the 
consequence of viral infections such as hepatitis, and after 
pregnancy. Other types of aplastic anemia are termed 
agranulocytosis or thrombocytopenia to indicate that the 
major deficiency lies in particular white cells or in 
platelet production, respectively. Agranulocytosis may be 
associated with autoimmune syndromes such as systemic lupus 
erythematosis (SLE) or with infections, particularly neonatal 
rubella. 

The overall mortality of all patients with aplastic 
anemias, in the absence of stem cell therapy, is high. 
Approximately 60-75% of individuals suffering from the 
disorder die within 12 months, in the absence of new stem 
cells. The overall incidence of these diseases is 
approximately 2 5 new cases per million persons per year. 
Although it is extremely unlikely that a single pathogenic 
mechanism accounts for all aplastic anemias, it is clear that 
provision of new hematopoietic stem cells is usually 
sufficient to allow permanent recovery, since transplantation 
of patients with aplastic anemia with bone marrow obtained 
from identical twins (i.e. , syngeneic) (Pillow, R.P., et al., 
1966, N, Engl, J, Med, 275 (2) : 94-97 ) or from HLA-identical 
siblings ( i , e, , allogeneic) (Thomas, E,D,, et al,, Feb. 5, 
1972, The Lancet, pp, 284-289) can fully correct the disease. 
However, some patients with aplastic anemia reject the 
transplanted marrow. This complication is particularly 
common among patients who have been immunologically 



35 




-58- 



sensitized as a result of multiple therapeutic blood 
transfusions. In a preferred embodiment of the invention 
employing autologous neonatal stem cells for hematopoietic 
reconstitution, such a complication can be avoided. 

In a specific embodiment of the invention, hematopoietic - 
reconstitution by infusion of neonatal stem cells can be used 
for the treatment of Fanconi's anemia, an autosomal recessive 
disease exhibited by congenital malformations associated with 
bone marrow failure. The stem cell defect is associated with 
chromosomal instability, and increased risk for malignancy. 
The disease is always fatal in its natural course. This 
embodiment of the invention is illustrated by way of example 
in Section 12, infra , which describes the infusion of 
neonatal blood comprising hematopoietic stem and progenitor 
cells into a patient with Fanconi's anemia for treatment of 
the disease. In a preferred aspect of this embodiment, the 
patient is conditioned before stem cell infusion, by a 
conditioning regimen which is modified according to cell 
sensitivity to alkylating agents and to irradiation ( see 
Gluckman, E. , et al. , 1983, Brit. J.. Haematol. 54:431-440; 
Gluckman, E. , et al., 1984, Seminars in Haematol. 21(1): 20- 
26; Gluckman, E. and Dutreix, J., 1985, The Cancer Bulletin 
37 (5) : 238-242 ; Gluckman, E. , et al., 1980, Brit. J. Haematol. 
45:557-564; all incorporated by reference herein). For 
example, cytogenetic analysis can be used to predict cell 
sensitivity to alkylating agents (Berger, R. , et al., 1980, 
Brit. J. Haematol. 45:565-568). Tests for radiosensitivity 
have also been described (Gluckman, E. and Dutreix, J., 1985, 
The Cancer Bulletin 37 (5) : 238-242 ; Gluckman, E. , et al., 
1983, Brit. J. Haematol. 54:431-440). In a particular 
embodiment, a conditioning regimen using cyclophoshamide and 
thoraco-abdominal irradiation can be employed. 



35 



5? 



-59- 



5.6.2, HEMATOPOIETIC MALIGNANCIES 

Hyperprol iterative malignant, stem cell disorders which 
can be treated by hematopoietic reconstitution with neonatal 
stem and progenitor cells include but are not limited to 
acute lymphocytic leukemia, chronic lymphocytic leukemia, 

5 

acute and chronic myelogenous leukemia, multiple myelomas, 
polycythemia vera, agnogenic myelometaplasia , Waldenstrom's 
macroglobulinemia, and Hodgkins and non-Hodgkins lymphoma. 
These leukemias are currently treated by chemotherapy and, 
when feasible, allogeneic bone marrow transplantation. 

10 

However, allogeneic HLA identical sibling bone marrow is 
available only to less than one-third of patients, and this 
treatment is associated with transplantation-related compli- 
cations such as immunodeficiency and graft versus host dis- 
ease. Provision of syngeneic (self) cryopreserved hemato- 

1 5 

poietic stem cells, according to a preferred embodiment of 
the invention, would permit hematopoietic reconstitution of 
patients lacking suitable allogeneic donors and eliminate the 
risks of graft versus host disease arising from allogeneic 
marrow transplantation. 

20 

5.6.3. MALIGNANT, SOLID TUMORS OF 
NON-HEMATOPOIETIC ORIGIN 

Hematopoietic reconstitution can greatly aid in the 

treatment of patients with malignant, solid tumors undergoing 

25 irradiation or chemotherapy, by providing new stem cells. 
Such tumors include but are not limited to those listed in 
Table II, supra . 

There is increasing evidence that a number of cancers 
are remarkably sensitive to extremely high doses of normally 

30 ineffective anti-neoplastic drugs. These cancers include 
malignant melanoma, carcinomas of the stomach, ovary, and 
breast, small cell carcinoma of the lung, and malignant 
tumors of childhood (including retinoblastoma and testicular 
carcinoma), as well as certain brain tumors, particularly 



-60- 

glioblastoma. However, such intensive high dose chemotherapy 
is not widely used because it frequently causes hematopoietic 
failure and death. The provision of new stem cells after 
intensive chemotherapy has been accomplished by using bone 
marrow cells obtained from patients befc_*e administration of 
the cytotoxic drugs (Spitzer, G. , et al., 1980, Cancer 
45:3075-3085). This approach has two major difficulties. 
First, it has not been routinely possible to obtain 
sufficient numbers of bone marrow cells from chronically ill 
patients with cancer. In addition, clinicians have been 
reluctant to use this approach because of the probability 
that the patient's bone marrow cells are contaminated by 
small numbers of neoplastic cells. This is particularly true 
in the hematologic malignancies, but also pertains to most 
metastatic cancers. The provision of stem cells according to 
the present invention, obtained at a time of health, before 
the onset of cancer, can permit the use of potentially 
curative intensive chemotherapy without the risk of stem cell 
failure. 

5.6.4. AUTOIMMUNE DISORDERS 
Many chronic inflammatory and degenerative diseases are 
characterized by a continuous immune reaction against the 
body's own tissues. Such autoimmune disorders include but 
are not limited to rheumatoid arthritis and other inflam- 
matory osteopathies, diabetes type I, chronic hepatitis, 
multiple sclerosis, and systemic lupus erythematosus. 
Autoimmune disorders are often treated by lymphoid irradi- 
ation. Use of the neonatal hematopoietic stem and progenitor 
cells for hematopoietic reconstitution according to the 
present invention can be extremely valuable after radio- 
therapy. 

Anti-inflammatory drugs such as steroids retard the 
inflammatory cells which are activated by autoreactive T 
cells, but do not prevent T cells which recognize self- 




10 



15 



20 



25 



30 



35 



-61- 

proteins from activating new inflammatory cells. A more 
direct approach to treating autoimmune diseases depends on 
eradication of T cells by irradiation of the lymphoid 
tissues, and relying on stem cells from the unirradiated bone 
marrow to repopulate the patient ' s hematopoietic system. The 
rationale is that the formation of new populations of mature 
T cells from bone marrow stem cells may result in absence of 
T cells that have reactivity to self-specific antigens. This 
procedure, called total lymphoid irradiation (TLI) , has been 
used to treat intractable rheumatoid arthritis (Strober, S., 
et al., 1985, Annals of Internal Medicine 102:441-449, 450- 
4 58) . These clinical trials showed that in the majority of 
otherwise intractable cases, joint disease was significantly 
alleviated for at least 2-3 years. However, the major 
drawback to such treatment is failure of stem cells in the 
bone marrow of these elderly patients to efficiently 
repopulate the hematopoietic sytem, resulting in infections 
and bleeding disorders. Analogous studies have been made of 
the effects of TLI as an alternative to cytotoxic drugs for 
treatment of SLE (Strober, S., et al., 1985, Ann. Internal 
Med, 102:450). Studies of the use of TLI to treat 
intractable SLE have also shown that this treatment 
alleviates disease activity, but is severely limited by 
failure of bone marrow stem cells to rapidly and efficiently 
repopulate the hematopoietic system after irradiation. In a 
preferred aspect of the invention, the availability of an 
individual's own stem cells, obtained at birth, can allow 
efficient repopulation of mature T cells in an adult 
environment, after minimal lymphoid radiotherapy, and can 
thus render this therapy significantly more effective. 

5.6.5. GENE THERAPY 
Hematopoietic reconst itution with the neonatal stem and 
progenitor cells of the invention which have undergone gene 
therapy, i.e. , which have stably incorporated a heterologous 




-62- 



gene capable of expression by their progeny cells, can be of 
great value in the treatment of diseases and disorders 
affecting cells of hematopoietic lineage * In one embodiment, 
hematopoietic reconstitution with such recombinant stem cells 
can be used in the treatment of genetic disorders of the 
hematopoietic system. Such genetic disorders include but are 
not limited to those listed in Table II, supra . Genetic 
deficiencies or dysfunctions of hematopoietic cells can be 
treated by supplying, to a patient, recombinant stem and 
progenitor cells. In a specific embodiment, patients who 
have hematopoietic cells which lack a gene or have a mutant 
gene, can be reconstituted with neonatal stem and progenitor 
cells that have incorporated a functional counterpart of the 
deficient gene. In particular, such genes which can be 
subject to gene therapy include but are not limited to 
hemoglobin or enzymes which mediate its synthetic pathway 
( e.g. , for treatment of anemias such as beta-thalassemia, 
sickle-cell disease) . 

In another specific embodiment, patients with infections 
by pathogenic microorganisms which occur in or affect a 
hematopoietic cell lineage can be treated with recombinant 
neonatal stem and progenitor cells. Such recombinant stem 
and progenitors can contain a heterologous gene which is 
expressed as a product which ameliorates disease symptoms, is 
toxic to the pathogen without significant detriment to the 
host, or interferes with the pathogen's life cycle, etc. 
Pathogens which cause infections which may be treated with 
recombinant stem cells according to this embodiment of the 
invention include but are not limited to lymphotropic viruses 
such as Human Immunodeficiency Virus (HIV, the etiological 
agent of acquired immune deficiency symdrome (AIDS)) (Gallo 
et al., 1984, Science 224:500-503; Barre-Sinoussi , F. , et 
al., 1983, Science 220:868; Levy, J. A. , et al., 1984, Science 
225:840); gram-negative bacilli such as Brucella or Listeria ; 
the mycobacterium which cause tuberculosis, or which cause 




10 



15 



20 



25 



30 



35 



-63- 

Hansen's disease (leprosy) ; parasites such as Plasmodium (the 
etiological agents of malaria) , or Leishmania ; and fungi 
(such as those that cause pneumonia and other lethal 
infections secondary to immunodeficiencies) (for a discussion 
of many of these disorders, see Harrison's Principles of 
Internal Medicine, 1970, 6th Edition, Wintrobe, M.M., et al., 
eds., McGraw-Hill, New York, pp. 798-1044). As a particular 
embodiment, it is possible to construct recombinant neonatal 
stem or progenitor cells that express a sequence which is 
"anti-sense* to the nucleic acid of a hematopoietic cell 
pathogen. Such a sequence, which is complementary to the 
pathogen's RNA or DNA, can hybridize to and inactivate such 
RNA or DNA, inhibiting the function or expression of the 
nucleic acid and disrupting the pathogen's life cycle. As a 
particular example, recombinant neonatal hematopoietic cells 
can be used in the treatment of AIDS, a disorder which is 
caused by HIV, apparently by infection of T4 + lymphocytes 
(Dagleish et al., 1984, Nature 312:763-766; Klatzmann et al., 
1984, Nature 312:767-768). Recombinant neonatal stem and 
progenitor cells which express an anti-sense nucleic acid 
that is complementary to a critical region ( e.g. , the long- 
terminal repeat or polymerase sequence ) of the HIV genome 
(Wain-Hobson et al., 1985, Cell 40:9-17) can be used for 
hematopoietic reconstitution for the treatment of AIDS. 

Numerous techniques are known in the art for the 
introduction of foreign genes into cells and may be used to 
construct the recombinant neonatal hemapoietic stem and 
progenitor cells for purposes of gene therapy, in accordance 
with this embodiment of the invention. The technique used 
should provide for the stable transfer of the heterologous 
gene sequence to the stem cell, so that the heterologous gene 
sequence is heritable and expressible by stem cell progeny, 
and so that the necessary developmental and physiological 
functions of the recipient cells are not disrupted. Tech- 
niques which may be used include but are not limited to 




-64- 



chromosome transfer ( e.g. , cell fusion, chromosome-mediated 
gene transfer, micro cell-mediated gene transfer) , physical 
methods ( e.g. , transf ect ion , spheroplast fusion, microin- 
jection, electroporation, liposome carrier) , viral vector 
transfer ( e.g. , recombinant DNA viruses, recombinant RNA 
viruses) etc. (described in-Cline, M.J., 1985, Pharmac. 
Ther. 29:69-92, incorporated by reference herein). 

5.6.6. MISCELLANEOUS DISORDERS 

INVOLVING IMMUNE MECHANISMS 

10 Hematopoietic reconstitution with the neonatal hemato- 

poietic stem and progenitor cells of the present invention 
can be used to treat patients with various miscellaneous 
disorders involving immune mechanisms. Disorders resulting 
from inefficient function, lack of function, or dysfunction, 

1 5 of an hematopoietic cell lineage can be alleviated by 

replacement of the hematopoietic cell progeny with those 
derived from neonatal stem and progenitor cells of normal 
function. In a specific embodiment, a hemolytic disorder can 
be treated (for a discussion of hemolytic disorders, see 

20 e.g. , 1985, Cecil, Textbook of Medicine, Wyngaarden, J.B. and 
Smith, L.H., eds . , 17th Ed., W.B. Saunders Co., pp. 900-915). 
Hemolytic disorders acquired during adult life account for 
the large majority of this form of anemia, and reflect the 
destruction of red cells by lymphocyte products. Stem cell 

25 replacement therapy with the neonatal cells of the invention 
can provide a new source of red cells, and, in an embodiment 
employing autologous cells, can replace destructive 
lymphocytes with newly formed cells which are unlikely to 
generate an immune response against the recipient's red 

30 cells. In another specific embodiment, patients whose immune 
system is compromised e.g. , as a result of irradiation or 
chemotherapy, can be treated by hematopoietic reconstitution 
with neonatal hemapoietic stem and progenitor cells (see 
Section 5.6.3). In yet another embodiment, disorders 

35 




-65- 



involving disproportions in lymphoid cell sets and impaired 
immune functions due to aging can be treated by 
reconstitution with the neonatal cells of the invention. 
Genetic disorders of metabolism which result in pathologic 
accumulations of metabolic products in the marrow ( e.g. , 
osteopetrosis, metabolic storage diseases) are also among the 
many disorders envisioned for treatment. 

In addition , immune deficiencies which are the primary 
or secondary result of infection by pathogenic microorganisms 
can be treated by hematopoietic reconstitution with the stem 
cells of the invention. In this embodiment, neonatal stem 
cells can serve as a source of cells of the hematopoietic 
cell lineage which are needed by the patient. For example, 
immune deficiencies caused by microorganisms which are 
intracellular pathogens of hematopoietic cells, can be 
treated by the provision of new hematopoietic cells, supplied 
by infused neonatal stem cells. Microorganisms causing 
immune deficiencies which may be treated according to this 
embodiment of the invention include but are not limited to 
gram-negative bacilli such as Brucella or Listeria , the 
mycobacterium which are the etiological agents of 
tuberculosis or of Hansen's disease (leprosy) , parasites such 
as Plasmodium (the etiological agents of malaria) or 
Leishmania , and fungi (such as those that cause pneumonia and 
other lethal infections secondary to immunodeficiencies) (for 
a discussion of many of these disorders, see Harrison's 
Principles of Internal Medicine, 1970, 6th Edition, Wintrobe, 
M.M., et al., eds., McGraw-Hill, New York, pp. 798-1044). 



In another method of the invention, progeny cells of 
hematopoietic stem and progenitor cells of fetal or neonatal 
blood can be generated in vitro ; the differentiated progeny 
cells thus generated can be therapeutically useful. For 



30 



5.7 



GENERATION AND USE OF HEMATOPOIETIC 
STEM AND PROGENITOR CELL PROGENY 



35 




-66- 



example, in one embodiment of this aspect of the invention, 
hematopoietic stem cells and/or CFU— GEMM progenitor cells, 
before or after cryopreservation and thawing, can be induced 
to differentiate into platelets. Such platelets can be used, 
for example, for infusion into a patient with 

5 

thrombocytopenia. In another embodiment, granulocytes can be 
generated in vitro prior to infusion into a patient. One or 
more of the hematopoietic progeny cells can be generated in 
vitro , allowing for the in vitro production of blood com- 
ponents. In a preferred embodiment, the generation of 
10 differentiated blood components is accompanied by expansion 
of the hematopoietic stem and progenitor cell pool, in order 
to allow for production of a greater quantity of differen- 
tiated cells. Various growth factors can be used to promote 
expansion and/or differentiation of hematopoietic stem and 

15 

progenitor cells, such as cytokines (growth factors) 
including but not limited to G-CSF, CSF-1, IL-3 , IL-5, tumor 
necrosis factor-a, and 7-interf eron. The blood components 
which are thus produced have uses which are not limited to 
therapeutic uses in vivo . For example, such progeny cells 

20 

can be used in vitro , e.g., for the production and isolation 
of hematopoietic cell products such as growth factors, 
antibodies, etc. 

6. EXAMPLES 

25 

6.1. COLLECTION OF HUMAN UMBILICAL 
CORD AND PLACENTAL BLOOD 

Neonatal blood was collected from human umbilical cords 

by gravity drainage and/or by needle aspiration from deliv- 

30 ered placentas. Data for the volumes obtained in one series 

of collections from individual births is shown in Figure 1, 

and demonstrates that volumes of 50 ml or more can be 

obtained. Data from another series of collections is shown 

in Figure 2, with the collections from individual births 

35 




# • 

-67- 



10 



15 



20 



30 



identified by method of collection and delivery type, as 
either: gravity flow, vaginal delivery; gravity flow, 
Caesarian section; placental aspiration, vaginal delivery;, or 
placental aspiration, Caesarian section. The data show that 
the majority of the collections had a total volume of greater 
than 30 ml although many contained less than 50 ml. In 
recent collections, we have been able to obtain volumes 
approximately twice as large as shown in Figure 2 ( e.g. , 99 
ml blood from a neonate, after a 36 week gestation) by using 
needle aspirations from the delivered placenta, at the root 
of the placenta and in the distended surface veins, combined 
with cord drainage. 

Cord blood collections were done essentially as 
described in Section 5.1.1 and subsections, supra , and as 
detailed infra. 



Cord blood collection kits consisted of: 

wide-mouth bottle (200 ml) (Corning, Corning, NY) , 
Cat. No. 25625-200; VWR, South Plainfield, 
NJ, Cat. No. 28199-756) 
wrap (operating room drape sheet) 
For collections by needle aspiration, 60 cc syringes B-D 
Luerlok (VWR, Cat. No. BD5663) and 18 gauge needles 1 1/2 
inch (VWR, Cat. No. BD5196) were used. 

Collection bottles were sterilized before collection by 
beta-irradiation with 2.5 megarads from a tungsten source 
25 (Dynamatron Accelerator, Radiation Dynamics, Inc., Melville, 
New York) „ Syringes and needles were autoclaved. (Alterna- 
tively, the syringes and needles were sterilized with ethy- 
lene oxide. ) 

Twenty ml of CPD (citrate-phosphate-dextrose) was added 
to each cord blood collection container, as an anti- 
coagulent. CPD was prepared according to the following: 
Trisodium citrate (dihydrate) 28.8 g 

Citric acid (monohydrate) 3.2 g 

Sodium dihydrogen phosphate 2.19 g 





10 



15 



-68- 

( monohydra t e ) 
Dextrose 2 5 . 0 g 

" Bring volume to 1,000 ml; pH should be 5,63. Use 

at 2 0 ml CPD per up to approximately 120 ml blood. 
In selected samples, acid-citrate-dextrose (ACD) (Hurn, 
B.A.L., 19 68, Storage of Blood, Academic Press, New York, p. 
137) was used instead of CPD. ACD was prepared according to 
the following: 

Trisodium citrate (dihydrate) 1,65 g 

Citric acid (monohydrate) 1.983 g 

Dextrose (anhydrous) 6.13 g 

in a total volume of 2 50 ml 
The substitution of ACD for CPD caused no observable 
differences in the hematopoietic stem and progenitor cell 
counts which were obtained. 

Penicillin and streptomycin were also added to the 
collected blood. 0.01 X cord blood volume, of a solution 
consisting of 5000 units penicillin per ml and 500 ug 
streptomycin per ml, was added to each cord blood sample. 

Approximately 109 of the human umbilical cord blood 
samples which were collected were subjected to further 
analysis as described infra . 

6.2. HEMATOPOIETIC STEM AND PROGENITOR 
CELLS IN COLLECTED CORD BLOOD 

25 The approximately 109 collected cord blood samples of 

section 6.1 were sent by overnight mail (in polystyrene 
mailers; Fisher, Fairhaven, New Jersey, Cat. No. 03-528-10) 
to a processing site where they were separated, counted for 
viable cell numbers, set up for hematopoietic progenitor cell 

30 assays (in most cases), frozen away for storage, and in some 
cases defrosted for assessment of recovery of total nucleated 
cells and hematopoietic progenitors (see Section 6.7, infra ) . 
The progenitor cells evaluated included immature and mature 
granulocyte-macrophage (day 7 CFU-GM, day 14 CFU-GM) , 

35 



20 




-69- 



"immature* and "mature" erythroid (BFU-E-1, BFU-E-2) , and 
multipotential cells (see Section 6.6 and subsections, infra 
for assays of progenitor cells) . Table III presents a 
complete list of the samples received and the numbers of 
hematopoietic progenitor cells per sample present in the low 
density fraction after separation with Ficoll-Hypaque. 



10 



15 



20 



25 



30 





10 



15 



20 



25 



en 
I 

O 
01 t—i 
» — I 

*-H X 

<U 

O en 
<D 

U -w 

-u o 

-H rH 

§8 

cT to 
v-< 03 

«-H <U 

03 CO 

4-> to 

O 0) 

E-* U 



05 -Q 

S3 



CQ 



CM 
I 

w 
I 



CQ 



9 



•9 

-H 
> 



p m 



03 
Q 



+j *n toi 

-«H Q i — I O 

1)1 O H H 

^ CQ U X 



(fi O H 
C • — I <D — * 
g CQ U 



30 



< — I -p 
(13 03 

S3 



1 

to o 



<D x 



35 



D £2 U — ' 
2 



0) 



CO 



O «— i cm 
on r- 

LO 



VD rr 

r- m vo 



VDOO CO 
rH O 

• — I oo 



ON CO ON 
IT) VD H 
H VD H 



O ^ m 
■^r VD (M 



03 
C 



c 
o 
u 

U 



to 
0) 

4J 
rH 

u 



a, 



to 
to 



<u 



T3 
03 

x: 



CO 



03 



03 



< — l > — < CM 



CQ 



m m \o 
o LO 



a = 

vo en m on -P cm »x> co on 

H CO H ri : CM CO 
rH tO rH 

-P 



(n vo to co 

r- cm on 



co co lO 

Sro o 
= rH rH 

rH 

§■ 



t o n oo n 4-J invDco o 
n <n en <n -h rH r- 



■a 

> 



VO O O ON rH rH 

H H n di r 

u 



on in o 

LO CM 



a 
o 



<D 

to 



MDOOONHHTr^VOCOvO O 
O ON I — H OJ (N C 



io\DOino>inootrHr»in 



a 

<U 
CO 

-p 

n n O oo 
i — i CM o c o 

H ^ H rH 
I 



in n Q) 

n in w 
on on on on 

o 
c 

I 

H fN on T3 vo (\r-TC0fNJ0003Oi — n 
co on on (Drr'OinoNr*niO(\iOHnooN 

vd vd ^ (N cl) co rHrnrHcomrrCJNvOrn^-t 

CM 03 > - — I * * 

CD -H 

rH <D 

a: 

in o m O-o OrH^^vomocor^rMrH 



i,o m id io n 

ON ON ON ON ON 



(N VD H (N n 
ON ON ON ON 



ai 



U 

-P 

to 

-P 

o 



CJ 
03 



03 



CQ 



m 

CM 



in o 
cm 



CM 

in 



in 

CM 



o 

ON 



Csl 

co 



o 
in 



CO 
CO 



m ko 
cm on 



r\i i co 03 cm 
r-~ vo vo CO co 



rH r- m 04 m 
m nin Mn 



rH CM CM CO 
LO I — VO 



CO 



ro rn 
on on 



rn o 
co ro 
cm 



ro in 
■^r in 



h f\ n in \o i — a) on <h , — i 
I i i I I I I i I i I 

CQpQCQOQCQOQCQCQaaCQtD 
UOUOUUOCJUUU 



(Nn^rm\Of^oocNC-i(N 

^— ( ,-— ( ^ ^-j ^-H 4 i CM CM CM 

1111(111111 

CQ£DQ£aC3CCQC3£3C2C2 

uuuuuuuauuu 



CM 
CO 



on 



oo 

ON 



CM 

I 

CO 

u 




-7 1- 



10 



15 



o 

to *-h 

— « x 

CJ W 

O C 

-U O 

•H «-H 

c o 
a. 

m w 

g g 



20 



25 



0) 

> 



E 

U 



w 

E 

CQ 



Ol 
I 

CO 

E 

CQ 



2 r- 

£03 
Q 
U — 



>, vo 

4J ■n «i 

-•-< Q «— ( O 

(0 0 H H 

CQ U X 



8 



4J 



<0 



^ CQ CJ 



"8 

« — i jj 

C 0J 

4-» 0) 



VO 

I 



30 



35 













O 








ni 

vU 








CD 
















03 
















J3 
















^3 
















<L) 








10 












oj 


co 


o 




co 




c 








>l 








<TJ 








10 








tO 








03 






ON 






, — ( 


co 


















0 
























05 






ON 


a 




CM 


<— 1 










« — i 








rH 
O 








U 






















co 






VO 


co 






^t* 




fte 








o3 

•a 






r- 






OJ 








«— I 






<— H 






U 
















to 








< — ( 








«-( 
















a 








CM 




co 








OO 




O 







ocr\or-ojco^oor-a\in 
• h cm n ao 

o 



ON Ol ON VO 
in r-t CO 



o vo o *-n 



vo 



co O ^ ON 

oi -rr vo oj 



n oj m n 
n r- oo (N 



-P 
CO 

O : 
C 



o vo o 



oj oj co on on m 
en (N n m h 



oj co on -w 

ro in oi -«^r « — i 



«— i on • a\ in n 
o « — i 



8- 

If) 



> 



oi^voioh 
vo on co 



ON «-H 
« — I OJ 



O vO 

co vo 



co in 



o on 

OJ I 



4~> 

o 
c 



T3 
<D = 



o o co o h 



oj h n OO CO O OJ 
cxi • oj ro h (N in 
o 



CO r 



«— i in o in 
m i— n co 



co 



co on -^r vo o 



+ 

•O f\ m vo ^ 

0) vd CN <N 

Qj co co 



co rr 

^ ON 



r-~ ro moNinr^r^o^vovo^ri — ONr-<— tr^voovommvor-*^r 
rsi^H inorovovom'crinr^r-tmr-vD Q)r-in^innH(N^m 

ONCVJ ONONONONONONONON ON COONCOONCJONONONONr^inmONON 



to 


o 




VO 


>—< 




m 


CO 


ON 


r- 


^* 


OJ 


VO 


o 


ON 


OJ 


o 




CO 


ON 


CO 


ON 


CO 


ON 




ON 


VO 


»— 1 




in 


o 


CO 


o 


r- 


CO 


CO 


ON 


VO 




OJ 


OJ 


r- 


. — ( 


<— i 


< — 1 


1— ( 




ON 




co 


CO 


CO 


.—I 


CO 


*— 1 




ON 


< — \ 


r-H 


CO 


CO 


CO 


«— 1 


in 






rr 


XT 


in 


CO 


VO 






CO 




VO 


CO 










<D 


X 






































« — 1 


OJ 












U 


























































o 


m 


OJ 


o 




CO 


m 


CO 






o 


VO 


in 




o 


o 


in 


VO 


o 


o 


in 


o 




VO 


VO 




i 


CO 




o 


o 


CO 


CO 


ON 


vo 




CO 


1 


VO 


in 


VO 


VO 


1-4 


Ol 


vO 




VO 


OJ 


CO 




r- 


• — I 


o 




\£> 


CM 






CO 


CO 


OJ 




OJ 


co 




OJ 


CO 


m 


in 


Ol 


Ol 


in 


» — 1 


CO 


in 




*— ( 


Ol 


CO 


> 








































« — i 




















in 


vO 


r- 


CO 


o 


o 


»— \ 


OJ 


CO 


CO 




tn 


VO 




CO 


ON 


o 


r-i 


OJ 


CO 




m 


vo 










rs) 


r\j 


OJ 


CM 


OJ 


ro 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 










"^r 


t 


l 


•^r 
1 




1 




1 

CQ 


1 

CQ 


i 

CQ 


1 

CQ 


1 


1 

CQ 


1 

CQ 


1 

CQ 


CQ 


1 

CQ 


1 

CQ 


1 

CQ 


1 

CQ 


l 

CQ 


I 

CQ 


1 

CQ 


I 

CQ 


1 

CQ 


I 

CQ 


t 

CQ 


1 

CQ 


CQ 


CQ 


CQ 






c 


U 


U 


CJ 


U 


CJ 


U 


U 


u 


U 


u 


u 


CJ 


CJ 


CJ 


CJ 


U 


CJ 


U 


CJ 


CJ 


CJ 


CJ 


U 


U 



0(1 



-72- 



10 



15 



co 
i 

o 
tn «-h 
« — i 

rH X 
<U 

cj cn 

0) 

u *H 

o c 

-P O 
•H rH 

C O 

a) a 
8* tn 
a. 

<-t Id 
03 in 
-P to 

e § 



20 



g 



CQ 



CM 
I 

w 

1 

CO 



>1 

£(0 
a 



5 



^ ic 1 
E a 



25 



<— i i— ( 

(0 JQ 

rH 

■a 

-«H 
> 



•H Q » — i o 

01 _ 

c « — i <u 



i _ VO 

T3 0)1 
O rH f- 



tn 



W O H dP 
C rH <U — ' 
g CQ U 



30 



13 . vo 

d) Q h 

*— t O »—t 

a co cj — 



35 



0) 

CO 



03 

CP-rH 

D oo 

O CD 
V-l 

o 



3* 



tn <u 
a) 

r 

tn 



4-J 

a. 



U« O 

4-> -C 



u 

(D 

u 



h- vd in 

VO 



co 
co co rH 



O (M ON 

rH rH in 



in in 
♦ oo 

vO 



co in in 



m o co 

VDVOOO 



O •sD T 

co cn 

CO CM <T» 



r- vo o 



oo in in 

CM CM OO 



CO o 
in 

t i i 

CQ CO CQ 
U U U 



o co vo o in 
oj h oo m oo 



Mn n h 



\o n vo <Ti 
n rg ro 



r*- co oo in vo in r-* 

CO rH T <J\ <J\ CO rH 
rH 



O rH VO CO V-~ CO VO 

in co r*- ^j* vo <-h cd 
co 



vo in co 

CM O "^T 



O rH O 

co vo r* 

CN rH rH 



in cn vo cm r- 
co rH o r*- o 

in rH 



^TOOOOrH^TOOCO 

<N(NOinnnvoHTT 

rH CO 



r- rH 

co r- 

CM rH 



CO 
CM 
CM 



vo m 

CM 



co 



ror^coococncM^rHcnocMO 
r^rrinn^rrnvoNinnm^r 

VO rH CO rH rH 



r--vpvococooovovotninvococovo 
(NHinn^HHnmnHH tn 

CM rH 



cm o in 
cm vo 

CM rH CM 



CO O rH 
in CO rH 



i_n o « — i co 

rH CM <T\ CM 
CO rH CM 



co cn cm cn co m in 
n cr> o \£) h 



VO 



0) 

tn 

8 



cn rH in 
in vo 

CO CM CM 



oo tn 
co r— 



^(N^TINlN^COfNIHOOin^Tr 



S 



03 

C co co in 



cMcoincMvomcMincOrHOcncMvo t h n r-i 
cncncncncocncncncn oo ov in r- cn 03 co cn r*- 

c 
o 
u 

rH 

in cm co r— i o> i — (N o in in vo co 03 cn co cn 
"=TvomcnrHOrHrHO'*3 , mcocOrH CP(N n 
cMcMcor^cMcocMP-voinvor-vocM cocor^ 

rH rH CM 3 CM rH 



momo^rm^oocomoococo coin 

CM CM • 

*<^r ro o co cn cn h o in o vo n o co r-~ 

cMCMinvOrH'^r'^rKrcocMCMCMCMrH rH «— I 



rH CO 

cn cn 



in 
cm 



o o 



< — irgn^inor-coc>o< — irgn-^invDr-cocnOH 
LnuOLntnuoininininvovovovovovovovoovor^r- 
(iiiiiiiiii(iiiiiii)t 
GjCQCQCCQC2D3CQacQG3CQCQCQCQC3GCDCDCQ 

uuuSuuuuuuuouuououuoo 



-73- 



10 



o 
tn i 
• — i 

«— < x 

CD 

CJ cn 
Q> 
V-l -H 
O C 

-P o 

•H r- ( 

c o 

(D (J 



8 1 

Cu 



15 



O 

tn 

(U 



4-> 



20 




25 



<D 


>1 










•§ i 


) (0 OH 




5 c h <u 


2* 





30 



"8 

O H O «— 1 



D CQ U 
2: 



35 



in co on ri in cu 
(N 00 vd h in 01 

CM 

o 
c 



(N in t oj 



00 00 in in o vd 
i — 1 r — vo cm 00 o 

*— I rH VD 



co 
cm 



O VD 

«— i in 



r- 00 m 
«h co co o 00 
• — i i — i « — 10 



en = 
cn 

s 



o 

in cm 
m -U 

5 



c 

N : 
O 



a 

CD 

ro oo n «h 

4-> rH H (N 
O 

c 
tn 

WvoHco^rcovor- 1 on n r- t cn 00 o o 
(O^^HONinrivo \d vo I s - in h fNi 

r- 1 ^ 

>. 
c 
o 

»H 

o 
u 

hOCOH^TOCOnfvjOHOfvifMai 

4ir*inHinoN(N^ in in in vd <- i cm 
D rH m CO 
-D 



o < — 1 ^ 1^- 
^ n h n 

CO CM «— I 



o r- 



go m o 

O VO CM 



00 o r — cm 
in 



00 

CM 



cn 



c > a\ co o 
O in 00 co vd 

tM CO CO CM 

2 

cn 



d)lDin<L(NOOC^(NCO<r»H^OH^ 

CjvovoocoinoNCM h n in \d 

CM rH r— I 



0 




GO 


CO 




tn 


CO 


VD 


O 


CM 


in 


CO 


CO 


cn 


O 


0 


r- 


CO 


0 






cn 


CO 


0 


CO 


O 


co 


rH 


VD 


r-* 


CM 


r- 


r— i 


Cn 


VD 


O 


CO 


CM 


H 




0 


VD 


in 


rH 


cn 






CO 






CO 


rH 


CM 


in 


X 




*— 1 


CM 


rH 


<— 1 


r- 




rH 




CO 




rH 


rH 


co 


VD 


rH 


rH 


CO 




CM 




rH 


rH 


rH 








VD 


cn 


«H 


rH 


CM 


r- 


in 


in 




VD 


m 


CO 




in 


VO 


CO 


CM 


in 


in 


CM 


0 


VD 


m 


m 


cn 




<*> 


«H 


0 


in 


CO 


0 


r- 


CO 




CO 


Cn 




CO 




CO 


CM 


en 


OO 


VO 


rH 




0 


VO 


00 


in 


H 


vo 




cn 


r- 


r- 


r- 


ON 


Cn 


0 co 


CO 


00 


CO 


ON 


cn 


ON 


ON 


CO 


cr\ 


cn 


cn 


cn 


cn 


cn 


CO 


CO 


cn 


cn 


D 
1 

O 




VD 


VD 




CM 


0 


VD 


in 


C- 


CM 


cn 


in 




O 




0 


CO 


00 


CO 


m 


CO 


CO 


CM 


0 


0 


vD 


rH 


r-H 


O 


r- 


ON 


0 


r- 


CO 




VO 


CO 


cn 


VD 


co 


VO 


r- 


CM 


VO 


rH 


CO 


0 


cn 


VD 


m 




m 


cn 






CM 


CO 




0 


tn 




00 


r- 


O 


in 




CO 


VO 


in 




ON 


CO 


rH 


rH 


CO 


VO 


vD 


r- 


— t 




X 




«H 


* — i 


« — 1 


rH 


CO 








CM 




rH 




rH 


rH 


rH 








rH 


















CM 






m 


0 


0 00 


CM 






C- 


r- 


CO 


CM 


O 


0 


in 


in 


O 


0 


CM 


CM 


r- 


in 


CM 














0 










in 






























vD 




co 


rH 


CM 


O 


r— 1 


co 


0 


cn 


CM 


CM 


m 


m 


m 


O 


r- 


CO 


0 


VD 


CO 


r- 


rH 


VO 


CO 




vD 






VD 


VD 


VD 


CO 


<— 1 


CO 


VD 


in 


rH 




CO 




VD 


VO 


in 


CO 


VD 


CO 


CO 


m 


m 


m 


in 


CO 


vc 


le 


CM 


ro 


-^r 


in 


VD 


r- 


CO 


cn 


0 


rH 


CM 


CO 




m 


VD 


r- 


CO 


cn 


0 


rH 


CM 


co 




in 


VO 


r- 




r- 


r- 


r- 






r- 






CO 


CO 


CO 


CO 


CO 


CO 


CO 


00 


CO 


CO 


cn 
l 


cn 
l 


ON 
I 


cn 


en 


cn 


cn 
1 






CQ 


1 

CQ 


1 

CQ 


1 

CQ 


» 

CQ 


1 

CQ 


1 

CQ 


1 

CQ 


CQ 


CQ 


1 

CQ 


1 

CQ 


1 

CQ 


i 

CQ 


CQ 


1 

CQ 


1 

CQ 


i 

CQ 


CQ 


CQ 


CQ 


CQ 


CQ 


CQ 


CQ 


CQ 


to 


O 


CJ 


a 


O 


CJ 


CJ 


CJ 


CJ 


U 


O 


U 


a 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


O 


O 


CJ 


CJ 




-74- 



o 

10 r-H 



10 



5 



15 



to 

. <U 
u 

o c 

4-> o 

c o 
a, 

h a) 

<o to 

-u (/) 

O (D 

H Jh 



20 



£ a 



>4 
a 




25 




4-> 'O (0 

«H O «— H - — ■ 

w (5 h op 

C «-H <L> — ' 

^ CQ U 



"8 



30 



<0 (0 *a 
4-> CU Q 
O —I O 



vo 
f 



P CQ U — 




35 



CO 



a= ========= 

CU= = z: = rr = = = 
0) 

in n o 

0= = = = = = = = = = 

c 

to 
>. 

(0= = = = = = = = = = 

O O « V£> 

h n A3 
>i 

o 

,-H=r = = r = = = = = 
O 
O 

o r- in 
h n iJ cm 

3 '- " " " '- - " " " 
3= ======== = 

CN d) ^ 
N= = r = = = = = = = 

o 
1-1 



CO 



t — i I <u 

u 



CO 



<n a\ n o aDtnocn*— i^r^r 
cm og cm » — i cm < — i «-h 



VO 

m 



<j\CTv0^o^r-cooOQ> cr* oo in cri (Ti 



to 


O 






oo 




o 


rn 




o 


oo 






CO 




CM 


«— 1 


« — ( 


in 




in 


m 


vO 




» — < 


CM 


ro 


VO 


r-H 


m 


CM 


o 








cm 


o 


r—t 




«— ! 




vO 


CO 


VO 


CM 




r- 


in 


QJ 


X 






*— < 








r— » 


r— < 


rn 


( 











omoornoNinoofNornino 

C>CO(NOOHn\0^'00^'OCT»r s '(N 



COONOOOOOOOOOO^-!^ 
CT^ CT* <— I «-™f *-H *-H «— * —~ «-H < «— ' r-H •— < 

i i i t i i i i t i t i i i 

uuuuuuuuuuuuuu 



10 



-75- 

From receipt, of the samples until the cells were frozen, 
16 hours were spent in processing the cells. (This time 
period included cell separation on Ficoll-Hypaque, counting 
the cells, setting up the progenitor cell assays, and freez- 
ing the cells) . 

As shown in Table III, significant total numbers of 
progenitors cells were obtained even with the overnight tran- 
sit time plus 16 hour processing- Even among the samples in 
transit for 48 hours, viable progenitor cells were observed. 
There was variability among donors in the observed number of 
progenitor cells. It should be noted that the values shown 
in Table III represent remaining progenitor cells after loss 
of progenitors due to cell separation procedures (see Table 
IV, infra ) . 

15 6.3. ENRICHMENT FOR HUMAN HEMATOPOIETIC 

STEM AND PROGENITOR CELLS: CELL 
SEPARATION PROCEDURES 

In a preferred embodiment of the invention, whole 
neonatal blood can be cryogenically preserved, and used for 
hematopoietic reconstitution after thawing, in order to avoid 
cell losses associated with cell separation procedures. 
However, it is envisioned that cell separation procedures can 
be used if desired, e.g. , to minimize blood storage volumes. 
Thus, in the examples sections herein, cell separation 
procedures are described which can be used to enrich for 
neonatal hematopoietic stem cells in collected blood. Many 
of the procedures relate to the enrichment of stem cells 
derived from adult bone marrow or adult blood, however, it is 
envisioned that the same procedures, or modifications there- 
of, are equally applicable to the neonatal hematopoietic stem 
cells of the present invention. 

Human stem and progenitor cells are present in the non- 
adherent, low density, T-lymphocyte-depleted fraction of bone 
marrow, spleen, and (adult and cord) blood cells. Purifica- 
tion or enrichment for the stem and progenitor cells has been 

35 



20 



25 



30 



-76- 



carried out by Ficoll-Hypaque density separation, adherence/ 
non-adherence separation, and positive selection by antibody 
binding. 



10 



15 



20 



25 



30 



6.3*1- DENSITY SEPARATIONS . 

Enrichment for human hematopoietic stem and progenitor 
cells has been carried out by isolation of low density 
(density less than 1.077 gm/cm 3 ) cells separated by Ficoll- 
Hypaque (Pharmacia Fine Chemicals, Piscataway, NJ) . 

The following protocol is used for samples of bone 
marrow or peripheral blood: 

1. Obtain sample of bone marrow or peripheral blood. 
Bone Marrow - sample should be 1-5 ml containing 

heparin. Place in sterile 17 X 100 mm 
tube. Add 2-3 ml of sterile DPBS 
(phosphate-buffered saline without 
magnesium or calcium). Mix well. 
Whole Blood - Dilute sample at least 1:1 with McCoy's 5A 
medium or DPBS. Adjust volume to a 
multiple of 20 ml. 

2. Bone Marrow - spin for 10 minutes at approximately 400 X 

g (1500 rpm; Beckman TJ-6R rotor) at 4°C. 
Whole Blood - Go to next step. 

3. Bone Marrow - Remove buffy coat and wash once with DPBS. 

Resuspend to a final volume of 20-40 ml 
(if 1-2 pulls of 2 ml each, resuspend to 
20 ml; if 3-5 pulls, resuspend to 40 ml). 
Count cells; adjust volume to a maximum of 
6 X 10 7 cells per 20 ml. 
Whole Blood - Go to next step. 

4. Bone Marrow - With a 10 ml pipet, carefully layer 20 ml 

of buffy coat suspension onto 15 ml of 
Ficoll-Hypaque in a 50 ml polypropylene 
tube. 



35 



Whole Blood - With a 10 ml pipet, carefully layer 20 ml 
of blood suspension onto 15 ml Ficoll- 
Hypaque in a 50 ml polypropylene tube. 

5. Using a balance, carefully adjust weight of tube(s) with 
blank(s) - 

6. With slow acceleration, centrifuge sample (s) at 400 X g 
(1500 rpm) for 3 0 minutes at 4°C. Turn brake off. 

7. Carefully remove the low density band and place it in a 
clean sterile tube. Dilute at least 1:10 with McCoy's 5A 
medium or DPBS. 

8. Wash cells twice by centrif ugation at 400 X g for 10 
minutes at 4°C. Resuspend to 50 ml and repeat. 

9. Final resuspension should be to 10-15 ml with McCoy's 
media. 

10. Perform cell count. 

The following modification of the above procedure has 
been used for cord blood separations (and was used in 
obtaining the data shown in Tables III and IV) : 

1.. Obtain cord blood, aseptically, using a 60 cc syringe 
containing 3 000-4000 units of preservative-free sodium 
heparin or ACD as an anticoagulant. 

2. Perform low density cell separation using Ficoll-Hypaque 
density gradient, by diluting cord blood with sterile DPBS 
(phosphate buffered saline without Mg ++ , Ca ++ ) , pH 7.0, at a 
ratio of 1:3 (cord blood: PBS). Layer 20 ml of blood 
suspension on 15 ml of Ficoll-Hypaque in a 50 ml polypropy- 
lene centrifuge tube (Fig. 3). Centrifuge at 4°C, 400 X g, 
for 30 minutes. 

3. Collect and pool all low density cell bands (Fig. 4) 
from each individual donor. Make sure that very little 
Ficoll-Hypaque is collected with cells, or the cells may not 
pellet through the collected Ficoll. Dilute the cell 
suspensions 1:1; if "X" ml of cells were collected, dilute 
with "X" ml of DPBS in order to dilute collected Ficoll 



-78- 



sufficiently to allow cells to pellet. Pellet cells by 
centrifugation at 4°C, 200 X g, for 10 minutes. 

4. Aspirate and discard supernatant from each pellet. If 
several tubes were used, pool identical donor pellets after 
resuspending each pellet with 5 ml of DPBS. Pellet cells by 
centrifugation at 4°C, 200 X g, for 10 minutes. 

5. Aspirate and discard supernatant. Resuspend pellet in 
10 ml of DPBS using a 10 ml pipet and gentle up-down 
aspirations. Bring volume to 50 ml with DPBS. Pellet cells 
by centrifugation at 4°C, 200 X g for 10 minutes. 

6. Resuspend in RPMI-1640 medium supplemented with 5% 
autologous plasma or heat-inactivated fetal calf serum (FCS) . 
Perform cell counts and viability. Keep cell suspension 
chilled to 4 °C. 

The effect of various density separation procedures on 
the yield of progenitor cells in the human cord blood 
collected (described in Section 6.1, supra ) was assessed. We 
have compared the number of progenitors in whole blood, no 
separation treatment, to that of whole blood in which the 
mature erythrocytes were lysed by treatment with ammonium 
chloride (NH 4 C1) , low density cells after Ficoll-Hypaque 
separation (density less than 1.077 gm/cm 3 ) , heavy density 
cells after Ficoll-Hypaque separation, and heavy density 
cells after treatment with NH 4 C1 to lyse the erythrocytes 
(Table IV, Exp. 1) . 



25 



30 



35 




-79- 



10 



\J 25 



TABLE IV 

COMPARISON OF HEMATOPOIETIC PROGENITOR CELLS 
OBTAINED WITH DIFFERENT CELL SEPARATION PROCEDURES 

Progenitor Cells X 10~ 3 



Separation CFU-GM CFU-GM CFU- 

Procedure day 7 day 14 BFU-E-2 BFU-E-1 GEMM 

Exp #1 



None (Whole Blood) 


167 


220 


330 


356 


356 


Whole Blood + NH„C1 


55 


112 


43 


39 


43 


Low Density (Ficoll) 


35 


87 


49 


23 


36 


Heavy Density (Ficoll) 


49 


104 


71 


82 


153 


Heavy Density + NH 4 Cl 


17 


40 


17 


14 


11 


Exp #2 












None (Whole Blood) 


561 


1020 


612 


484 


408 


Whole Blood Sedimented 
with Methyl Cellulose 


157 


388 


212 


286 


111 


Low Density (Ficoll) 


256 


653 


506 


448 


186 


Heavy Density (Ficoll) 


3 


8 


8 


14 


5 


Exp #3 












Sample CB-57 

Whole Blood Sedimented 

with Methyl Cellulose 


6 


12 


11 


12 


6 


Low Density (Ficoll) 


35 


59 


30 


76 


37 


Sample CB-58 

Whole Blood Sedimented 

with Methyl Cellulose 


5 


14 


14 


17 


7 


Low Density (Ficoll) 


16 


30 


31 


43 


26 



35 



Sample CB-59 

Whole Blood Sedimented 



-80- 



with Methyl Cellulose 6 21 31 39 17 

Low Density (Ficoll) 13 52 64 67. 43 

Sample CB-60 

Whole Blood Sedimented 

with Methyl Cellulose 2 9 5 4 0 

Low Density (Ficoll) 3 40 18 18 6 



As shown in Table IV, Exp. 1, there are many more 
progenitors detected in the unseparated blood than in the low 

10 density Ficoll preparation. This difference is not due to 
loss of cells into the dense fraction of Ficoll, which 
contains mainly mature neutrophilic granulocytes. Lysing 
whole blood erythrocytes also resulted in a lower yield of 
progenitors. In experiment number 2, we compared whole 

15 blood, whole blood that was sedimented with methyl cellulose 
to remove erythrocytes, and low and high density Ficoll 
separated cells. The results demonstrated that whole blood 
contained the most progenitors, some of which were lost from 
the fraction of cells obtained after sedimentation of the 

20 er Y throc ytes with methyl cellulose. As seen in both 

experiments 2 and 3 of Table IV, sedimenting cells with 
methyl cellulose was inferior to the low density fraction of 
Ficoll with respect to numbers of progenitors. While the 
Ficoll separation removed mature granulocytes and erythro- 

25 c Y tes from the progenitor cell fraction, some progenitors 
were also lost, relative to whole blood, using this 
procedure . 

6.3.2. ADHERENCE/NON-ADHERENCE SEPARATION 
3q An adherence/non-adherence separation protocol for 

enrichment of hematopoietic stem and progenitor cells is as 
follows : 

1. In a 60 mm Corning tissue culture dish, seed 10-15 x 10 6 
low density cells in up to 3 ml of McCoy's 5A (supplemented) 
35 media with 10% fetal calf serum (heat-inactivated) . 






-81- 



2. Incubate for 1.5 hours at 37 °C in an atmosphere of 5% 
co 2 . 

3. Gently swirl plate to loosen non-adherent cells. Pipet 
into sterile centrifuge tube. Carefully rinse dish with 3 ml 
iXCoy's media and pool the media. 

4. Add 1 ml McCoy's media to the dish, and gently remove the 
cells with a sterile rubber policeman. Remove the cells and 
place them in a sterile centrifuge tube. Rinse the dish with 
3 ml media and pool media. 

5. Pellet cells by centrif ugation at 400 X g for 10 minutes 
at 4°C. Aspirate the supernatant and resuspend the cells in 
media. 

6 . Repeat step 5 . 

7. Perform cell count. 



The following protocol has been used for 
cryopreservation of viable hematopoietic stem and progenitor 
cells derived from human cord and placental blood: 
20 1- Pellet low density, Ficoll-separated cells by 
centrif ugation at 4*C, 2 00 X g for 10 minutes. 

2. Check viable cell count by trypan blue exclusion 
(Kuchler, R.J., 1977, Biochemical Methods in Cell Culture and 
Virology, Dowden, Hutchinson & Ross, Stroudsburg, Pa., pp. 

25 18-19) and manual cell counting using a hemocytometer . 

3. Gently resuspend cells to a concentration of 4 x 10 6 
viable cells/ml, using a mixture of cold (4°C) 50% autologous 
plasma/RPMI-1640 or 50% heat-inactivated FCS/RPMI, and place 
the suspension on ice. 

30 4 . In a cryovial containing 1 ml of a chilled, sterile 

cryoprotective medium of 20% DMSO/RPMI-164 0 , carefully layer 
a 1 ml portion of the aforementioned cell suspension on top 
of the cryoprotective medium. 



6.4. 



CRYOPRESERVATION OF CORD BLOOD 
STEM AND PROGENITOR CELLS 




-82- 



10 



15 



20 



25 



30 



5. Approximately 10 minutes prior to freezing, slowly invert 
the 1:1 mixture to promote mixing, then place it on ice to 
allow equilibrium between the cells and the cryoprotective 
medium. NOTE: The "layered" tube should not remain unfrozen 
for very, -long, so freezing should preferably be done within 
20-30 minutes after exposure of cells to DMSO/RPMI solution. 

6. Place the vials in a freezing rack, which in turn is 
placed in a 4°C methanol bath, just deep enough to cover the 
cell suspension (Fig. 4), This is then placed in the bottom 
(to ensure proper temperature) of a -80 °C freezer for at 
least 2 hours and less than 24 hours, 

7. After cells reach the frozen state, carefully and quickly 
transfer them to a long term liquid nitrogen containment 
vessel. A cryogenic storage vessel which can be used is the 
LR1000 refrigerator (Union Carbide Corp., Indianapolis, 
Indiana) which accommodates up to 4 0,000 cryules. 

6.5. CELL THAWING 
The following protocol has been used for thawing of 
cryopreserved cord blood stem and progenitor cells: 

1. Remove vial of frozen cells from liquid nitrogen. 
Immediately thaw cell suspension by gently agitating the vial 
in a 37 °c water bath until just a small amount of ice 
remains. 

2. Aseptically, begin to add drop-wise, a chilled mixture of 
50% autologous serum/RPMI-1640 medium or 50% FCS/RPMI-1640 
medium with a slight mixing between each drop, until the 
suspension volume is doubled. 

3. Transfer this suspension to a larger centrifuge tube 
(12-15 ml) and continue to add, drop-wise, 50% serum/RPMI 
mixture with mixing between every other drop until the volume 
reaches 6-7 ml. Diluent may now be added, drop-wise, with 
mixing at every 0.5 ml increment until the volume reaches 



35 




-83- 



9-10 ml. (NOTE: The reason for stepwise addition of diluent 
is to prevent osmotic shock to the cells as DMSO is diluted 
in the cell suspension.) 

4. Pellet cells by centrifugation at 4°C, 200 X g, for 10 
minutes. Aspira^a the supernatant. 

5. Slowly add, drop-wise, 1 ml of chilled 20% autologous 
serum/RPMI-1640 mixture to the pellet. 'Resuspend* the 
pellet by gently "flicking" the tube with a finger. After 
the pellet is resuspended (clumps may remain) , resuspend it 
further by gently aspirating up and down with a 1 ml pipet. 

6. Add an additional 4 ml chilled 20% autologous serum/RPMI, 
dropwise, with mixing between every drop; then add 0.5 ml as 
volume increases, as previously described. 

7. Pellet cells by centrifugation at 4°C, 200 X g, for 10 
minutes. Aspirate the supernatant. 

8. Resuspend with 2-5 ml of chilled 20% serum/RPMI mixture. 

9. Perform cell counts (by use of a hemocytometer) and 
viability testing (by trypan blue exclusion) . 

Loss of cells due to clumping during the stepwise 
removal of DMSO can be diminished by including DNase (20 U 
per 2 x 10 6 cells) or low molecular weight dextran and 
citrate (to reduce the pH to 6.5). 



Assays which can be used to quantitatively assess human 
hematopoietic stem and progenitor cells are described in the 
following examples sections. The assays for granulocyte- 
macrophage (CFU-GM) , erythroid (BFU-E) , and multipotential 
30 (CFU-GEMM) progenitor cells (Sections 6.6.1 and 6.6.2) were 
used to derive part of the data for human cord blood cells 
that is presented in Table III, supra . 



6.6. 



HUMAN HEMATOPOIETIC STEM AND 
PROGENITOR CELL ASSAYS 



25 



35 



10 



15 



-84- 



6-6.1. CFU-GM ASSAY 



The following assay has been used to quantify CFU-GM: 

1. Obtain a suspension of cells (cord blood, bone marrow, 
spleen, cell line, etc.) at a known cell concentration. The 
cell suspension concentration should be at least 10 fold 
greater than the final concentration desired in the plate. 

2. Depending on the number of plates to be plated, the 
volume of the culture mixture will vary. As an example, a 10 
ml suspension can be made, as described: 

In a 17 x 10 mm polystyrene tube, combine the following 
components except for the Agar (0.6%) and cells.* 

10 ml 

Agar (0.6% w/v) 5 ml (50%) 

(bacto-agar, Difco Corp.) 
**2X McCoys 5A 2 ml (20%) 

FCS (heat inactivated) 1 ml (10%) 

***Stimulator 1 ml (10%) 

♦Cells 1 m i (10%) 



* The cells are added just before adding the melted Agar, 
in order to avoid allowing the cells to sit in 2X McCoys 
for very long. Since the Agar has been boiled, care 
should be taken to allow it to cool sufficiently. 

** This volume may vary if the cells are more concentrated 
than desired. Example: if cells were to be plated at a 
final cell concentration of 1 x 10 5 cells/ml, and the 
stock cell suspension was 5 X 10 5 cells/ml instead of 1 x 
10 cells/ml, 0.2 ml cells would be used, plus 0.8 ml Of 
2x McCoys to achieve a 1 ml volume. In general, whatever 
volume is lacking after adding the other components, is 
made up with 2x McCoy's. (See Section 6.6.1.1, infra for 
the preparation of McCoy's medium) . 

*** Colony formation can be stimulated by factors present in 
medium conditioned by the 5637 urinary bladder carcinoma 
cell line (see Section 6.6.1.2 infra), which was used 



# • 

-85- 



routinely, or medium conditioned by the PHAL cell line 
(phytohemagglutinin-stimulated leukocytes from patients 
with hemochromatosis; Lu, L. and Broxmeyer, H.E., 1985, 
Exp. Hematol. 13:989-993), or by purified growth factors. 
Growth factors which may be Lasted for human colony 
stimulation include but are not limited to interleukin-3 
(IL-3) , granulocyte-macrophage (GM) -colony stimulating 
factor (CSF) , granulocyte (G)-CSF, macrophage (M) -CSF 
(also referred to as CSF-1) , erythropoietin, IL-1, IL-4 
(also called B-cell growth factor) and E-type 
prostaglandins. (These molecules are available in 
purified form from various companies, e.g. , Cetus, 
Immunex, and Amgen.) For murine cell assays, pokeweed 
mitogen spleen cell conditioned media may be used (see 
Section 6.6.1.3, infra ) . 

/ 

3. After the Agar has sufficiently cooled, add the appropri- 
ate volume of cells and 0.6% Agar. 

4. Place a cap on the tube, and mix the suspension well. A 
vortex may be used, but with caution. 

5. With an appropriate pipet, place 1 ml of the culture 
suspension into a 10 x 35 mm dish, containing colony 
stimulating factors if so desired. After all the dishes have 
been plated, allow them to solidify. 

6. Label the tray of plates and place it in the appropriate 
incubator. Incubation is conducted in a humidified atmos- 
phere of 5% C0 2 at low oxygen tension (5% 0 2 ) for 7 days and 
14 days. Low oxygen tension enhances the detection of CFU- 
GM, BFU-E , and CFU-GEMM cells. 

7. Remove plates from the incubator and score by 
observation of colonies under an inverted or stereoscopic 
microscope. Colonies scored at 7 and 14 days represent 
maturation stages of CFU-GM cells. (Day 7 CFU-GM represent a 
later or more mature progenitor of the granulocyte-macrophage 



-86- 



lineage, while day 14 CFU-GM represent an earlier progenitor 
of the granulocyte-macrophage lineage) . 



10 



15 



6.6.1.1. PREPARATION OF McCOY / S 5A MEDIUM 
The following procedure was used to prepare IX McCoy's 
5A Medium: 

1* 1 envelope McCoy's 5A medium (Gibco #430-1500) 



NaHCO, 



2.2 gm 



Bring to 1 liter with double-distilled H 2 0; pH to 7.0- 
7.2. 

2. Filter-sterilize by use of a 0.2 urn filter and peristal- 
tic pump (positive pressure) . 

3. If medium is to be used for growth or incubation, it is 
supplemented with the following: 



Per Liter 
of Media 




25 



8 ml 
4 ml 

10 ml 
4 ml 
10 ml 
15 ml 



MEM essential amino acids (Gibco #320-1130) 
MEM non-essential amino acids (Gibco #32- 
1140) 

MEM sodium pyruvate (Gibco #320-1360) 
MEM Vitamins (Gibco #320-1120) 
Penicillin-Streptomycin (Gibco #600-514 0) 
Serine/ Asparagine/Glutamine mixture (see 
recipe infra) 



To prepare 2X McCoy's 5A medium (for plating), follow 
the same procedure as for IX, except bring the volume only to 
500 ml instead of 1 liter. Add the same volume of 
30 supplements . 

The Serine/Asparagine/Glutamine mixture is prepared 

according to the following: 

L-asparagine (Sigma #A-0884) 800 mg 

L-serine (Sigma #S-4500) 420 mg 

35 





-87- 



L-glutamine (Gibco #320-5030) 200 ml 

1. Dissolve serine and asparagine in 450 ml double-distilled 
H 2 0) , bring the volume to 500 ml and filter-sterilize 
through a 0.2 urn filter 

2. Add to this sterile mixture 200 ml of L-glutamine. Mix - 
well and aliquot into 7.5 ml/tube. Store at -20 °C. 



10 



15 



20 



25 



30 



35 



6.6.1.2. PREPARATION OF HUMAN 5637 URINARY 
BLADDER CARCINOMA CELL LINE 
CONDITIONED MEDIUM 

The following procedure can be used to obtain medium 

conditioned by the human 5637 urinary bladder carcinoma cell 

line: 

1. Thaw and start cells from frozen stocks, per "Quick Thaw" 
protocol of Section 6.5, supra . Grow 5637 cells to 
confluence in a 150 cm 2 flask containing 50 ml of the 
following medium: 

RPMI 164 0 
glutamine (2 mM) 

penicillin-streptomycin (10 ml/liter at 

1000 units/ml) 
10% fetal bovine serum (heat-inactivated) 

2 . Incubate in an atmosphere of 5% C0 2 , with normal 0 2 , for 
3-5 days; check daily. 

3. Split the cells 1:20 into 20 X 150 cm 2 flasks with 50 ml 
RPMI 1640 media (as above) . 

4. Incubate for 7 days in 5% CO , normal O . 

5. At 3-5 days, if desired, select 1 flask of cells to 
prepare for freezing as a stock supply of cells. Freeze 10 6 
cells/vial (1 ml) for liquid nitrogen storage, per the 
protocol described in Section 6.4, supra . 

6. At 7 days, collect cell medium into 20 X 50 cc centrifuge 
tubes. Spin down cells and cell debris by centrifugation for 
10 minutes at greater than or equal to 500 X g. 





-88- 



7. Pool the medium, filter-sterilize it using a 0.2 um 
filter, and aliquot into 100 ml bottles. Store frozen at 
less than 0°C (usually -20°C or -80°C) . 

8. Assay stimulation activity of the medium by the CFU-GM 
assay described in Section 6.6.1, e.g. , using human marrov 

5 cells. 



6.6.1.3. PREPARATION OF MURINE POKEWEED MITOGEN 
SPLEEN CELL CONDITIONED MEDIUM 

The following procedure can be used to obtain murine 

10 pokeweed mitogen spleen cell conditioned medium (PWMSCM) , a 

crude source of growth factors for use in hematopoietic 

colony stimulation for mouse cells. 

1. Obtain a single cell suspension of CBA/J mouse spleen 
cells at a known cell concentration of greater than or equal 

15 to 20 x 10 6 cells/ml. (Mice should be 5-7 weeks old). 

2. Make a large volume of cell growth suspension as follows 



CBA/J spleen cells 2 x 10 6 cells/ml 

Heat-inactivated FCS 10% 
20 Pokeweed mitogen 0.333% (1:300) 

(Gibco #670-5360) 
Iscove's modified Dulbecco's Remainder 
media* + 3.024 gm NaHC0 3 
(Gibco #78-5220) 

25 

* Preparation described infra in Section 6.6.2.3. 
3. After mixing the above ingredients, place 50 ml of the 
mixture in a 50 cm tissue culture flask, and incubate for 7 
days at 37 'C in an atmosphere of 5% C0 2 . 
30 4. After seven days, collect the conditioned media and 

remove the cells by centrifugation at greater than or equal 
to 500 X g at 4°C for 10-15 minutes. 

5. Carefully remove the conditioned media from the tubes, 
and filter-sterilize the media by passage through a 0.4 5 um 



-89- 



filter. Store the conditioned media in 50 ml polyethylene 
tubes at -20°C. 



The following assays have been used to quantify BFU-E-2 
and BFU-E-1/CFU-GEMM. BFU-E-1 and BFU-E-2 are erythroid 
progenitor cells that are operationally defined, and are not 
proven to be physiologically distinct. The BFU-E-1 is 
operationally defined as an early erythroid progenitor cell 
capable of producing a colony of erythroid progeny cells in 
semi-solid medium, upon stiumlation by erythropoietin, hemin 
(optional) , and a burst-promoting factor* The BFU-E-2 is 
operationally defined as a more mature erythroid progenitor 
cell, capable of producing a colony of erythroid progeny 
cells in semi-solid medium, upon stimulation by 
erythropoietin and by hemin (optional) . BFU-E-1 colonies 
tend to be larger than BFU-E-2 colonies. 

For the BFU-E/CFU-GEMM assay, Iscove's modified 
Dulbecco's medium (IMDM) was used, with methyl cellulose as 
the semi-solid support medium. (This was in contrast to the 
CFU-GM assay, where McCoy's medium was used, with bacto-agar 
as the semi-solid support medium.) 

The procedure was the following: 

1. Obtain a single cell suspension of known concentration of 
the appropriate type of cells. 

2. Depending on the number of plates plated, the volume of 
the culture mixtures will vary. As an example, we will make 
a 3 ml mixture. In order to increase the mixture volume, 
simply increase component volumes proportionately. In a 17 x 
100 mm tube, mix the following components: 



6.6.2. 



BFU-E-2 AND BFU-E-1/CFU-GEMM ASSAY 



30 



3 ml 



Methyl cellulose (2.1%) 
Glutamine (200 mM, Gibco) 



30 



1.4 ml 



ul 



(2 mM) 



35 




-90- 



10 



— 2 

2-mercaptoethanol (10 M) 

(7 ul into 10 ml McCoy's 5A) 
*Hemin (4 mM) 
** Erythropoietin (20 units/ml) 
FCS (not heat-inactivated) 
Cells (at least 10 X desired) 
***Iscove's Modified Dulbecco's 

Medium (IMDM) 
***GM Stimulator (if desired) 



10 



ul 



75 ul 
0.15 ml 
0.9 ml 
0.3 ml 
0.135 ml 

0.01 ml 



(5 x 10 5 M) 



- (0.1 mM) 
(1 unit/ml) 
(3.0%) 



* Preparation described infra ; Lu, L. and Broxmeyer, H.E., 
1983, Exp. Hematol. 11 (8) : 721-729 . 



15 



20 



25 



**Note that there are different types of erythropoietin which 
can be used; as an example, Hyclone erythropoietin has been 
used in murine cell assays, and Toyobo erythropoietin has 
been used in human cell assays. Purified recombinant 
erythropoietin is commercially available ( e.g. , Amgen, 
Thousand Oaks, CA) and may be used. 

*** GM stimulators include but are not limited to various 
factors which can be tested for colony stimulation, as 
described for the CFU-GM assay. The volume of the GM 
stimulator, and thus of the IMDM, may vary with the type of 
stimulator used ( e.g. , mouse=PWMSCM; Human=5637 CM or 
PHALCM) . Also note that IMDM is strictly a compensation 
for the remaining volume of 3 ml. 



30 



35 



3. Mix suspension thoroughly by vortexing and inversion of 
tubes. 

4. After allowing bubbles to rise from the mixture, place 1 
ml mixture in each of two 10 x 35 mm culture plates 
containing erythropoietin, hemin, and colony stimulating 
factors, if so desired. Rotate the plates so that the 
mixture coats the surface of the plates. 



-91- 



10 



15 



20 



25 



30 



5. Place these 2 plates in a large 15 x 100 mm petri dish 
along with a 10 x 35 mm humidifying dish containing about 1 
ml 'of H 2 0. Replace the lid of the large dish. 

6. Place the petri dish in an appropriate incubator for 14 
days. Conditions of incubation are the same as described for 
the CFU-GM assay of Section 6.6.1. 

7. Remove plates from the incubator and score by observation 
of colonies under an inverted or stereoscopic microscope. 

In some cultures, the GM stimulator/burst-promoting 
activity ( e.g. , medium conditioned by 5637 cells or PHALCM) 
can be omitted; under these conditions, the assay detects a 
more mature population of BFU-E (BFU-E-2) cells and few or no 
CFU-GEMM cells. 

6.6.2.1. PREPARATION OF 2.1% METHYL CELLULOSE 
The 2.1% methylcellulose, for use in the BFU-E/CFU-GEMM 
assay, was prepared as follows: 



Stock solution ; 

2.1% Methocel (Dow Chemical Co.) 21 grams 

Boiling water 500 ml 

2x IMDM 500 ml 

Procedure : 



35 



The gram weight of methyl cellulose is put into a 
sterile 3 liter Erlenmeyer flask (having a sterile stopper) 
containing a sterile magnetic flea on a large magnetic 
stirrer. To prevent as little frothing as possible, stirring 
is initiated while 500 ml of sterile boiling distilled H 2 0 is 
gently poured down the sides of the flask. Stirring contin- 
ues at room temperature until the flask gradually cools (this 
may take an hour) . When the flask is no longer hot to the 
touch, 500 ml of 2X IMDM, which had been allowed to come to 
room temperature , is added to the flask without frothing. 
The flask is stoppered and transferred to the cold room (4°C) 




-92- 

where stirring continues for 48 hours. The solution is then 
sterilely aliquoted into sterile 100 ml bottles. The bottles 
are stored frozen for up to 6 months (protected from light) . 

6.6.2.2. PREPARATION OF HEM IN 
The hemin, for use in the BFU-E/CFU-GEMM assay, was pre- 
pared as follows: 

2 60 mg Hemin (Eastman Kodak #22 03) 
4 ml 0.5 M NaOH 

\y 5 ml Tris buffer, 1 M, pH 7 . 8 (approximately 

10 

9.5 parts acid to 3 parts base) 

Bring to 100 ml with double-distilled H 2 0. 

1. Dissolve hemin in NaOH completely before adding Tris 
buffer and H 2 0. 

2. After adjusting the volume to 100 ml, filter-sterilize by 
passage through an 0.45 um filter, and store in 2-3 ml ali- 
quots at -20 °C. 



15 



6.6.2.3. PREPARATION OF ISCOVE'S 

MODIFIED DULBECCO'S MEDIUM 

20 IX Iscove's Modified Dulbecco's Medium (IMDM) , for use 

in the BFU-E/CFU-GEMM assay, was prepared as follows: 
1. Measure out 5% less water (deionized, distilled) than 
desired total volume of medium, using a mixing container that 
is as close to the final volume as possible. 

25 2. Add powder medium (Gibco Laboratories, Formula No. 78- 
5220) , to water with gentle stirring at room temperature (do 
not heat water) . 

3. Rinse out the inside of the package, to remove all traces 
of the powder. 
30 4. Add 3.024 grams of NaHC0 3 per liter of medium. 

5. Dilute to the desired volume with water. Stir until 
dissolved. 

6. Do not adjust pH. Keep container closed until medium is 
filtered. 

35 





-93- 



7. Sterilize immediately by Nalgene filtration. 

To prepare 1 liter of 2X liquid medium, follow the above 
procedure, except use 2 envelopes of powder instead of one, 
and 6.048 gm NaHC0 3 . 

6.6.3. STEM CELL COLONY FORMING UNIT ASSAY 
The assay used for stem cell (S-cell) quantitation does 
not directly assay self -renewal, but instead assays for the 
ability to generate secondary multilineage colonies on 
replating. This assay is done essentially the same as the 
BFU-E/ CFU-GEMM assays, except that cultures are scored after 
21-28 days of incubation rather than after 14 days (for BFU-E 
and CFU-GEMM) . The drug 4-hydroperoxycyclo-phosphamide (4HC) 
appears to spare immature progenitors at the expense of 
mature progenitors, and may be useful for pretreating cells 
before assay. Factors which can be tested for increasing the 
self-renewal ability of S-cells in vitro (thus increasing 
assay efficiency) include but are not limited to hemin, 
oxygen tension (Smith, S. and Broxmeyer, H.E., 1986, Brit. J. 
Haematol. 63:29-34), superoxide dismutase, glucose oxidase, 
IL-3, GM-CSF, G-CSF, M-CSF, erythropoietin, IL-1, IL-4 , etc. 



6.6.4. ASSAY OF THE PROLIFERATIVE STATUS 
OF STEM AND PROGENITOR CELLS 

25 The proliferative status of stem and progenitor cells 

can be measured by a high specific activity tritiated 
3 

thymidine ( HTdr) kill (or suicide) technique, carried out as 
follows: 

1. In two small 12 x 75 mm polystyrene tubes, place the 
30 appropriate volume of stock cell suspension containing 2-3 
times the number of cells required for plating. (For bone 
marrow, 2-3 X 10 6 cells and for spleen, 15-20 X 10 6 cells. 
For cord blood: 2-3 x 10 6 (approx. ) cells.) Label them a and 
b. 



-94- 



2. Pellet the cells by centrif ugation at 200-400 X g at 4°C 
for 10 minutes. 

3. Carefully remove and discard the supernatant. 

4. Add 0.5 ml of McCoy's 5A medium supplemented as 
prescribed in Section 6.6.1.1, supra , and with FCS at 10% 
v/v. 

5. To tube b, add 50 uCi of 3 HTdr (New England Nuclear, 
#NET-027X Thymidine, [methyl- 3 H] -20 . 0 Ci/mmol; 5.0 mCi/5.0 ml 
H 2 0) . As a control, to tube a, add 50 ul of McCoy's 5A 
medium. 

6. Place cap back on tubes and gently vortex in order to 
resuspend cells. 

7. Place the tubes in a tray also containing H 2 0, in an 
incubator with an atmosphere of 5% C0 2 , and a temperature of 
37°C, for 20 minutes. 

8. Add 0.5 ml (2.5 mg) of ice cold (4°C) 'cold*' (nonradio- 
active) thymidine (Sigma #T-9250) at 5 mg/ml to each tube, 
and vortex lightly. Add an additional 2 ml of ice cold 
McCoy's 5 A medium to each tube. 

9. Pellet cells by centrif ugation at 200-400 X g at 4°C for 
10 minutes. 

10. Aspirate the supernatant into an appropriate container 
(one used for radioactive disposal) , and resuspend the cells 
with 2 ml cold medium. Repeat step #10. 

11. Aspirate the supernatant into an appropriate container. 
Resuspend with McCoy's 5A containing 10% FCS to a volume 
where the cell concentration is at least 10 fold greater than 
the plating concentration. 

12. Keep cells on ice until ready to plate. 

13. Plate and carry out colony forming assays as described 
supra in sections 6.6.1 through 6.6.3. 



35 




# • 



-95- 

6.7. RECOVERY AFTER FREEZE-THAWING 

OF HUMAN HEMATOPOIETIC PROGENITOR 
CELLS DERIVED FROM CORD BLOOD 

The results of progenitor cell assays after freeze- 

thawing were compared to results of the same assays obtained 

5 before f reeze-thawing, in order to assess the recovery of 
hematopoietic progenitor cells from human cord blood after 
the f reeze-thawing process. Eight cord blood samples, 
obtained as described in Section 6.1, supra , and separated by 
use of Ficoll-Hypaque, were analyzed. The results are shown 

n in Table V. 



15 



20 



25 



30 



35 



10 



15 



> 
3 



20 



25 



30 



0" 



8 



E 



B 

CQ 



CM 
I 

w 

PQ 



D Q 



1 

0) o 

rH rH 

*H 

> W 

5" 3 



CO CO 

o i 



CO 



co moo 
o o • 

rH CvJ oo 

n h n 



VO CM O 

cn h r- 



co o r- 

oo • 

co vo oo 
n h 



^ M H 

in vc 

(M H i — I 

in cm 



cm n 
co co • 
n (N h 

rH CM 



ro CN 



5 

03 

u o 

cu V-l 
<D 

-p 

03 



03 
> 



D 
CO 



o 
vo o 
oo cr\ o 
t cm 



o cm a\ 

VO • 

vo r- 

CM CM 



I 

CQ 
U 



t— i vo 

O rH • 

rH <J\ VO 

^ rH Tj« 



CO rH VO. 
CM VO • 
O CO CO 

cvj m 



^ co 

O CO • 

O VO 
VO CM 



CO CM C\J 
VO O • 
CM CT\ CO 

co rH in 



oo co 



vo vo in 
in cm • 
co co r— 
cm « — i 



tj- cm 
vo o • 
r- 

cm rH in 



CO CM rH 
O CO • 

n 
cm vo 



CM VO CO 
rH «T • 

h 

00 



CO 

VO CTi 

cr\ r- vo 

CM CM 



o cn VO 

CM CM 

rH OO VO 
CO CM 



VO *T 

tn cm 
ro in in 
n h tt 



o m r- 

o r- • 

CO rH O 

co 



CM 

TJ- O • 

rH O OO 
O CO CM 



-^r cm r- 
crv 

CO CO vo 

CO CM 



CM O C\ 

-. — t 

in r- oo 



cm o ^ 
in rr 

CT\ CM rH 



VO 

CO C\ • 
H VO 

r-H CM 



^ CM CO 

cm r- « 

^ V£) H 

co cm r*^ 



O 

O co 
O O T 

H H lA 



VO 
CO 



CO VO rH 

CO rH CM 



CTv CM CO 

CO rH O 



CO 
co 



00 
CM 



3 

<D W Ih 

J-( O 

Oi CU J-f 
<U 
-(J 

03 



03 
> 



3 
03 

Q) U) £ 
V-t O 
Oi Cu Jh 
<L> 

4J 
VM 

03 



03 
> 



5 
03 

<u to ^ 

^ O 

a a u 

(U 
.u 
vm 
03 



03 
> 



3 
03 

0J 10 f 

^ o 
a* a* u 



03 



CM 
I 

6 



CO 



co 
i 

CQ 
U 



CO 



CO 



03 
> 



Z3 
CO 



e 



i 

CQ 
O 



OO CM Tj* 

vo 

O CO 
CM i — i 



VO O CO 

o r-H r- 

CO rH CO 



O TT (N 
VO CM 

«— I CM 

CM CM r— I 



O VO CM 
CM CM 

a\ 
in 



co *-h r*- 



<U to 
u> O 



3 

03 



I 

CQ 

o 



VM 

03 



03 
> 



CO 



A 




-97- 



10 



15 - 

4J 

C 
O 

> 
I 

20 ^ 



25 



30 



u 
u c 

•rH CJ 

-U N 

0) o 

-H V-l 

ft," 

4J O 

S X 
MH 

o U 

u & 

a 
o 
c 

81 



2 



I 

w 

e 

CO 



CM 
1 

w 

I 

0Q 



03 



u-r <T3 
U O 



VO 
I 

0) o 

«— < rH 

"§ X 
> to 

s'3 





<u 




CP 




03 




V-i 


1 


s 






0) 


CO 


O 


1 


i 


eze 


a; 


<u 


j-i 


u 


a, 





35 



o ro co 
rH o\ 



vo o o 

ro oo • 

o eg r- 

*—\ cm 



CM O CTk 
ON <Ti • 
in t-H f*— 
VO 



o oo ^ 

rH 

h in 



0O (N VD 

rH rH 

1/1 H H 



<~0 O « — i 
CM 



<u 
a, 








— ~» 






ON 






CO 










in 




ON «— 1 




r> i 


in cm in 


o 


• 


ro 


CO 








rH 












. — v 






in 














CM O O 


VO 


1 

vo 


ro vo 


• 


• 


ro co r* 




vo 


•-H CM 




CM 













' — 






• 












vo 
1 




oo 


^ 


ON 






in i — i oo 


in 


CO 


CM 




CM 


















CM 






« 






CM 






rH 


o o o 




rH 


in o 




1 


in rH 


in 


r- 




vo 








o 
























CM 


o o o 


vo 


• 

CO 


o 




CM 


rH rH 


in 


rH 




vo 


1 






CO 






rH 






CM 


















VO 


r- m in 


rH 








r- 


CO o m 


VO 


vo 


rH 


CO 


I 






in 






CO 






rH 

















3 


















03 










SO 


Th 


re 


OS 












a, 




a, 


a. 




















Jh 












<u 






<D 






























03 










er 


























rH 






rH 




VM 


















03 








> 






> 












•<H 






••H 




rH 














> 


<u 


03 














JH 


LP 


> 
















03 


-«H 








CO 


rH 




CO 


J-i 






CP 












<U 












e 






3 


Su 




a: 



-98- 



As shown in Table V, the average % survival after freeze- 
thawing was 36,1, 65.6, 65.4, 45.8, 44.6, and 30.5, respect- 
ively, for nucleated cells, day 7 CFU-GM, day 14 CFU-GM, 
BFU-E-2, BFU-E-1, and CFU-GEMM. There was a range of varia- 
bility in recovery rates. 

It should be noted that the data presented in Table V 
reflects cell losses incurred during Ficoll-Hypaque 
separations and procedures for DMSO removal, two steps which 
are omitted in a preferred embodiment of the invention (NB: 
DMSO should be removed before colony assays if such are 
10 desired to be carried out) . 

6.8. CALCULATIONS OF THE RECONSTITUTING 
POTENTIAL OF CORD BLOOD 

The following discussion demonstrates that individual 
15 collections of cord blood (such as described in Section 6.1) 
contains sufficient hematopoietic stem and progenitor cells 
to repopulate the hematopoietic system of an individual. 

A survey of published reports indicates that the number 
of CFU-GM infused for autologous bone marrow reconstitution 
20 in human patients, can be relied on as an indicator of the 
potential for successful hematopoietic reconstitution 
(Spitzer, G., et al., 1980, Blood 55(2): 317-323; Douay et 
al., 1986, Exp. Hematol. 14:358-365). By standardizing 
published data by patient weight, and assuming a patient 
25 weight of 150 pounds (67.5 kilograms), the calculated number 
of CFU-GM needed for successful hematopoietic reconstitution 
using autologous bone marrow cells ranges from 2-425 x 10 4 , 
with faster recovery noted using greater than 10 x 10 4 CFU- 
GM. 

30 The data presented in Table III, supra , for 81 cord 

blood collections, analyzed for day 14 CFU-GM count, shows a 
range of 0-109 x 10 4 CFU-GM per Ficoll-Hypaque-separated 
individual blood collections. Seventy samples contained 
greater than or equal to 2 x 10 4 CFU-GM, while thirty samples 

35 




10 



15 



20 



25 



30 



-99- 



as 



contained greater than or equal to 10 x 10 4 CFU-GM. It 
should be emphasized that this data is derived from Ficoll- 
Hypaque-separated cells obtained by either gravity drainage 
from the cord or needle aspiration from the delivered 
placenta. In a preferred embodiment of the invention, where 
vhQle blood is both frozen and infused for therapeutic use, 
losses due to Ficoll-Hypaque separation can be avoided (see 
Table IV and Section 6-3.1 infra for data on cell losses 
incurred during Ficoll-Hypaque separations). In addition, 
mentioned in Section 6.1, supra , in recent blood collections, 
we have been able to obtain volumes approximately twice as 
large as shown in Figure 2 or described in Table III, by 
using needle aspirations from the delivered placenta at the 
root of the placenta and in the distended surface veins, in 
combination with cord drainage. Furthermore, an adjustment 
of the collection protocol to provide for immediate cord 
clamping upon delivery should result in receipt of greater 
blood collection volumes (See Yao, A.C., et al., October 25, 
1969, Lancet: 871-873, wherein collected neonatal blood, 
obtained by drainage from the umbilical cord and from the 
delivered placenta, averaged 126.6 ml volume when the 
umbilical cord was clamped in less than 5 seconds after 
birth) . Thus, although an analysis of the data of Table III 
should be adjusted for expected losses during f reeze-thawing 
(which losses, however, should not exceed 35%), there should 
be sufficient cord stem and progenitor cells per collection 
sample to successfully effect hematopoietic reconstitution. 
Furthermore, the reconstituting capacity of cord blood 
hematopoietic cells may be higher than that of an equal 
number of bone marrow cells. Colonies derived from cord 
blood cells are usually larger in size than those derived 
from adult bone marrow. 



35 




-100- 



6.9. IN VITRO CULTURE CONDITIONS FOR 

HEMATOPOIETIC STEM AND PROGENITOR CELLS 

Culture conditions for the growth in vitro of 

hematopoietic progenitor cells from human cord blood have 

been described in Smith, S. and Broxmeyer, H.E., 1986, 

British Journal of Hematology, Vol. 63, pp. 29-34, which is 

incorporated by reference herein in its entirety. Culture 

media was composed of the following ingredients: 

RPMI 1640 media (Gibco Laboratories, Grand Island, NY) 
10 6 M hydrocortisone (Sigma, St* Louis, MO) 
5 ug/ml Vitamin D 3 (U.S. Biochemical Corp., Cleveland, 
OH) 

20% fetal calf serum, heat-inactivated (Hyclone 
Laboratories, Logan, UT) 

2 gm/1 NaHC0 3 (Fisher Scientific Co., Fair Lawn, NJ) 

100 U/ml Penicillin 
100 ug/ml Streptomycin 
0.2 5 ug/ml Fungizone 

Various conditions and factors can be tested for any 
effect increasing the self-renewal ability of stem cells in 
vitro. These include but are not limited to oxygen tension 
(see Smith and Broxmeyer, 1986, Br. J. Hematol . 63:29-34, 
incorporated by reference herein) , superoxide dismutase 
(Sigma Chemical Co., St. Louis, Mo.), glucose oxidase (Sigma 
Chemical Co.), and combinations of various colony stimulating 
factors, namely interleukin-3 (IL-3) , granulocyte-macrophage 
(GM) -colony stimulating factor (CSF) , granulocyte (G)-CSF, 
macrophage (M) -CSF (CSF-1) , erythropoietin, IL-1, and IL-4 (B 
cell growth factor) . 

6.10. MOUSE DISSECTION PROTOCOLS 
Mouse bone marrow and spleen are valuable sources of 
murine hematopoietic stem and progenitor cells for model 



-101- 



studies testing new and/or improved protocols for use with 
the human neonatal stem and progenitor cells of the present 
invention. Procedures for dissection of mouse bone marrow 
and spleen are described in Sections 6,10,1, and 6.10.2, 
respectively. 

6.10.1. BONE MARROW DISSECTION 
The following procedure can be used to obtain a murine 
bone marrow cell suspension: 

1. Sacrifice mouse as prescribed by cervical-thoracic 
dislocation. 

2. Inside a laboratory hood, soak the mouse with 70% ethanol 
(to avoid microbial contamination) , completely wetting 
the fur. 

3. Snip through the skin, and peel the skin down to the hip 
by holding the foot with either forceps that have been 
soaked in 7 0% ethanol, or with fingers, and pulling the 
skin with forceps. 

4. With sterile (alcohol-treated) forceps and scissors, cut 
away as much muscle tissue as possible to expose the 
femur. 

5. Remove the tibia from the femur by cutting through the 
knee cartilage/ joint . Discard the tibia. 

6. Remove the femur from the body by placing the sharp edge 
of a scissors on the anterior side of the hip joint, and 
pulling the femur in the opposite direction against the 
scissors, so that the scissors fits in the fold. Snip 
through the joint. 

7. Remove the knee end of the femur first, by snipping just 
the end with a scissors. Remove the hip end from the 
femur by the same method. 

8. With a 10 cc syringe containing 5 ml media (McCoys 5A IX) 
and a 27 gauge needle, place the needle in the bone cav- 
ity via the hip end of the bone. 



-102- 



10. 



15 



20 



25 



30 



9* Flush the marrow from the bone by forcing media into the 
cavity with the syringe, while holding the bone and 
syringe over a 17 x 100 mm tube, 

10. After both femurs have been evacuated, break up clumps 
with a 10 cc syringe and a 23 gauge needle. 

11. Pellet the cells by centrifugation at 400 X g (1500 rpm 
in a Beckman TJ-6R rotor) for 10 minutes at 4°C. 

12. Aspirate the supernatant and discard it. 

13. Resuspend the cells with 10 ml McCoys 5A media and a 
pipette, and repeat steps 11 and 12. 

14. Resuspend the cells with 10 ml McCoy's 5A media with a 
pipette, and count the cells (with a hemocytometer) . 

6.10.2. SPLEEN DISSECTION 
The following procedure can be used to obtain a murine 
spleen cell suspension: 

1. Sacrifice mouse as prescribed by cervical-thoracic dis- 
location. 

2. Inside a laboratory hood, soak the mouse with 70% ethanol 
(to avoid microbial contamination) , completely wetting 
the fur. 

3 . Place the mouse on its abdomen and snip through its 
left side skin with a sterile scissors and forceps. 

4. Lift the peritoneum with the forceps, and snip through to 
the abdominal cavity. 

5. With the spleen in view, remove it and place it in a 60 X 
100 mm dish containing 5-7 ml media. 

6. Place the spleen in a sterile homogenizing screen, in the 
dish, and snip it into small pieces. 

7. With the plunger of a 10 cc syringe, gently work the 
tissue through the screen into a dish containing media. 

8. Transfer the cell suspension from the dish to a tube. 
Rinse the plate with 3 ml media and pool. 





-103- 



9. Resuspend small pieces by transferring the cell 
suspension from the tube to a 10 cc syringe, and passing 
it through a 23 gauge needle twice . 

10. Pellet the cells by centrif ugation at 400 X g (1500 rpiu) 
for 10 minutes at 4*C. 

5 

11. Aspirate the supernatant and discard it. 

12. Resuspend the cells with 10 ml McCoy's 5A media and a 
pipette, and repeat steps 10 and 11. 

13. Resuspend the cells with 10 ml McCoy's 5A media, and 
count the cells (with a hemocytometer) . 

6.11. HEMATOPOIETIC RE CONST I TUT I ON OF 
ADULT MICE WITH SYNGENEIC FETAL 
OR NEONATAL STEM CELLS 

The experiments described in the examples sections infra 
15 demonstrate the hematopoietic reconstitution of adult mice 
with syngeneic or Tla-congenic stem cells of fetal or 
neonatal blood. 

A key reference and source of citations for use in 
animal model studies, which describes standards for experi- 
2Q mental irradiation, of mice and other mammals, at the level 
causing 100% mortality from hematopoietic failure, and 
prevention of such mortality by hematopoietic reconstitution 
(with bone marrow cells), is: Balner, H. Bone Marrow 
Transplantation and Other Treatment after Radiation Injury, 
25 Martinus Nijhoff Medical Division, The Hague, 1977, which is 
incorporated by reference herein. 

6.11.1. HEMATOPOIETIC RECONSTITUTION OF LETHALLY- 
IRRADIATED MICE WITH STEM CELLS 
IN BLOOD OF THE NEAR-TERM FETUS 

30 The examples herein described demonstrate that stem 

cells in blood of the near-term fetus are able to reconsti- 
tute the hematopoietic system of lethally-irradiated mice. 

The irradiated mice were ten (B6 X A-Tla b )F ]L hybrid 
males, aged seven weeks. The mice were exposed to 862.8 rads 



-104- 



at a radiation dose of 107.85 rad/min for 8 minutes with a 
137 

Cs source. This dose constitutes the LD100/30 days, i.e., 
the minimum or near-minimal Lethal Dosage causing 100% 
mortality within a 30-day post-irradiation period. Use of 
the 3 0-day survival endpoint is standard because hematopoie- 
tic reconstitution is deemed sufficient by that time, and any 
later mortality is therefore attributable to causes other 
than hematopoietic failure. 

Blood was collected from five near-term (B6-Tla a X A) 
hybrid fetuses, delivered by Caesarian section from one 
mother. In this experiment, near-term fetuses were used 
instead of neonates in order to ensure microbial sterility. 
The genetics of donor and recipient mice provides complete 
histocompatibility except for a segment of chromosome 17 
bearing the Tla marker gene. All mice were maintained 
previously and throughout on acidified drinking water to 
eradicate pseudomonas and similar infective organisms. 

As a restorative treatment, three mice each received 
0.17 ml heparinized whole fetal blood (made up to a total 
volume of approximately 0.2 ml by adding M199 medium with 
penicillin and streptomycin added) by intravascular injection 
into a peri-orbital vein of the eye, within two hours of 
irradiation. The results (Table VI) demonstrated the 
resultant survival of mice reconstituted with fetal blood 
stem cells, in contrast to the observed death of mice which 
had undergone no restorative treatment. 



30 



35 




-105- 



TABLE VI 

HEMATOPOIETIC RECONSTITUTION OF LETHALLY- 
5 IRRADIATED ADULT MICE WITH STEM CELLS 

IN BLOOD OF THE NEAR-TERM FETUS 

3 0-day Sur- 

Group Day of Death vival Rate* 

(1) Treated 14 2/3** 

10 (2) Controls; no restorative 11, 12, 12, 0/7 
treatment but conditions 13, 13, 15, 
otherwise identical 15 



15 



20 



25 



30 



* All 3 0-day survivors were normally healthy over prolonged 
periods of observation, displaying the typical post- 
irradiation graying of the coat, and would doubtless have 
experienced an approximately normal life-span, as is 
typical of reconstitution with syngeneic or near-syngeneic 
cell donors* 

** Later typing for the Tla marker by cytotoxicity assay of 
thymocytes (Schlesinger , M. , et al., 1965, Nature 
206:1119-1121; Boyse, E.A., et al., 1964, Methods in 
Medical Research 10:39) established repopulation by donor 
cells of the injected blood. 



6.11.2. HEMATOPOIETIC RECONSTITUTION OF MICE 

WITH A LESSER VOLUME OF NEAR-TERM FETAL 
BLOOD BUT NOT WITH ADULT BLOOD 

The examples herein described demonstrate that a defined 
volume of near-term fetal blood contains adequate hematopoi- 
etic stem cells to effectively reconstitute the hematopoietic 
system of lethally-irradiated mice, while the same volume of 
adult blood will not effect successful reconstitution. 



35 



10 



15 



20 



# 



-106- 



The irradiated mice were 20 (B6 x A-Tla ) F hybrid males 
aged 7 weeks, and 10 (B6 x A-Tla ) F females aged 7 weeks. 
The mice were exposed to 862.8 rads at a radiation dose of 
107.85 
days) . 



13 7 

107.85 rad/min for 8 minutes with a Cs source (LD100/30 



Blood was collected from eight near-term (B6-Tla x A) F^ 
hybrid fetuses, delivered by Caesarian section from one 
mother. In this experiment, near-term fetuses were used 
instead of neonates in order to ensure microbial sterility. 
The genetics of donor and recipient mice provides complete 
histocompatibility except for a segment of chromosome 17 
bearing the Tla marker gene. All mice were maintained 
previously and throughout on acidified drinking water to 
eradicate pseudomonas and similar infective organisms. 

As a restorative treatment, 10 mice received 0.02 ml 
heparinized whole fetal blood per mouse (made up to a total 
volume of 0.22 ml by adding M199 medium with penicillin and 
streptomycin added) , and 10 mice each received 0.02 ml adult 
whole blood identically treated, by intravascular injection 
into a peri-orbital vein of the eye, within 2 hours of 
irradiation. Control mice received no restorative treatment. 
The results (Table VII) demonstrated that stem cells in a 
defined volume of fetal blood can successfully reconstitute 
the hematopoietic system, while cells in an equal volume of 
adult blood cannot. 



25 



30 



35 




-107- 



10 



15 



30 



35 



TABLE VII 

SUCCESSFUL HEMATOPOIETIC RECONSTITUTION 
WITH A DEFINED VOLUME OF NEAR-TERM 
FETAL BLOOD BUT NOT WITH ADULT BLOOD 

30-day Sur- 

Group Day of Death vival Rate* 

(1) Treated with fetal blood 10,12, 12, 5/10** 

14, 14 

(2) Treated with adult blood 11, 11, 12, 0/10 

12, 12, 13, 

14, 14, 15, 
15 

(2) Controls: no restorative 9, 10, 10, 0/10 

treatment but conditions 11, 11, 12, 

otherwise identical 12, 12, 15, 

23 



* All 30-day survivors were normally healthy over prolonged 
periods of observation, displaying the typical post- 
20 irradiation graying of the coat, and would doubtless have 
experienced an approximately normal life-span, as is 
typical of reconstitution with syngeneic or near-syngeneic 
cell donors. 

25 ** Later typing for the Tla marker by cytotoxicity assay of 
thymocytes (Schlesinger , M. , et al., 1965, Nature 
206:1119-1121; Boyse, E.A., et al., 1964, Methods in 
Medical Research 10:39) established repopulation by donor 
cells of the injected blood. 




-108- 

6-11.3. HEMATOPOIETIC RECONSTITUTION WITH 
BLOOD OF NEWBORN MICE IN VOLUMES 
AS LOW AS TEN MICROLITERS 

The examples herein described demonstrate that the stem 
cells in a volume of neonatal blood as low as 10 microliters 
5 can reconstitute the hematopoietic system of lethal ly- 
irradiated mice. 

The irradiated mice were 20 (B6 x A-Tla b )F 1 hybrid males 
aged 8-12 weeks. The mice were exposed to 862.8 rads at a 
radiation dose of 107.85 rad/min for 8 minutes with a 137 Cs 
1Q source (LD100/30 days) . 

Blood was collected by cervical section from eighteen 
(B6-Tla a x A)F 1 hybrid neonates, less than 24 hours old. As 
a restorative treatment, 5 mice received 0.04 ml heparinized 
whole neonatal blood per mouse (made up to a total volume of 
15 approximately 0.2 ml by adding M199 medium with penicillin 

and streptomycin added), (Group 1); 5 mice each received 0.02 
ml (Group 2); 5 mice each received 0.01 ml (Group 3); and 5 
mice received no further treatment (Group 4, radiation 
control) . Treatment was by intravascular injection into a 
2Q peri-orbital vein of the eye. 

The genetics of donor and recipient mice provides 
complete histocompatibility except for a segment of chromo- 
some 17 bearing the Tla marker gene. All mice were main- 
tained previously and throughout on acidified drinking water 
25 to eradicate pseudomonas and similar infective organisms. 
The results in Table VIII show that stem cells in 
extremely small neonatal blood volumes (down to 10 ul) were 
able to reconstitute the hematopoietic system. 



35 



/b? 



10 



15 



-109- 



TABLE VIII 

SUCCESSFUL HEMATOPOIETIC RECONSTITUTION 

WITH NEONATAL BLOOD VOLUMES 
AS LOW AS TEN MICROLITERS 

30-day Sur- 

Group Day of Death vival Rate* 

(1) Treated with 0*04 ml 12 4/5** 
neonatal blood 

(2) Treated with 0.02 ml 14, 18 3/5 
neonatal blood 

(3) Treated with 0.01 ml 12, 12, 14, 1/5 
neonatal blood 14 

(4) Controls: no restorative 5, 6, 9, 10, 0/5 
treatment but conditions 11 

otherwise identical 



* All 30-day survivors were normally healthy over prolonged 
periods of observation, displaying the typical post- 
20 irradiation graying of the coat, and would doubtless have 
experienced an approximately normal life-span, as is 
typical of reconstitution with syngeneic or near-syngeneic 
cell donors. 

25 ** Later typing for the Tla marker by cytotoxicity assay of 
thymocytes (Schlesinger , M. , et al., 1965, Nature 
206:1119-1121; Boyse, E.A., et al., 1964, Methods in 
Medical Research 10:39) established repopulation by donor 
cells of the injected blood. 

30 



35 



/ID 



10 



15 



20 



25 




-110- 



6.11.4. HEMATOPOIETIC RE CONST I TUT I ON 
WITH BLOOD OF NEWBORN MICE IN 
VOLUMES OF 10 OR 15 MICROLITERS 

The examples herein described demonstrate that the stem 
cells in a volume of neonatal blood as low as 10 or 15 
microliters can reconstitute the hematopoietic system of 
lethally-irradiated mice. 

The irradiated mice were 15 male and 5 female (B6 x A- 
Tla )F 1 hybrids aged 10-12 weeks. The mice were exposed to 
862.8 rads at a radiation dose of 107.85 rad/min for 8 
minutes with a 137 Cs source (LD100/30 days) • 

Blood was collected by cervical section from fourteen 
(B6 x A-Tla b )F 1 hybrid neonates, less than 24 hours old. As 
a restorative treatment, 10 mice received 0.015 ml 
heparinized whole neonatal blood per mouse (made up to a 
total volume of approximately 0.2 ml by adding M199 medium 
with penicillin and streptomycin added), (Group 1); 5 mice 
each received 0.01 ml (Group 2); and the 5 female mice 
received no further treatment (Group 3, radiation control). 
Treatment was by intravascular injection into a peri-orbital 
vein of the eye. The donor and recipient mice were 
genetically identical, and thus completely histocompatible . 
All mice were maintained previously and throughout on 
acidified drinking water to eradicate pseudomonas and similar 
infective organisms . 

The results shown in Table IX reveal that stem and 
progenitor cells in neonatal blood volumes of 10 or 15 
microliters were able to reconstitute the hematopoietic 
system. 



30 



35 



ii 



-111- 



10 



15 



20 



25 



30 



TABLE IX 

SUCCESSFUL HEMATOPOIETIC RE CONSTITUTION 

WITH NEONATAL BLOOD VOLUMES 
OF 10 OR 15 MICROLITERS , 

30-day Sur- 

Group Day of Death vival Rate* 

(1) Treated with 0.015 ml 12, 12, 12, 4/10 
neonatal blood 13, 13, 13 

(2) Treated with 0.01 ml 12, 16 3/5 
neonatal blood 

(4) Controls: no restorative 12, 13, 14, 0/5 
treatment but conditions 17, 22 
otherwi se ident ica 1 



All 3 0-day survivors were normally healthy over prolonged 
periods of observation, displaying the typical post- 
irradiation graying of the coat, and would doubtless have 
experienced an approximately normal life-span, as is 
typical of reconstitution with syngeneic or near-syngeneic 
cell donors. 



35 



6.12. HEMATOPOETIC RECONSTITUTION FOR 
TREATMENT OF FANCONI 9 S ANEMIA 

In the example herein, we describe a procedure 
which was carried out to effect the hematopoietic 
reconstitution of a patient by allogeneic peripheral blood 
stem cell infusion, for treatment of the genetic anemia 
Fanconi ' s syndrome . 

The patient was a 5 year old white male* child with 
Fanconi 's anemia. The patient was first noted to be 
pancytopenic at 24 months of age. He was subsequently 
confirmed to have Fanconi 's anemia by diepoxybutane-induced 




-112- 



chromosomal breakage assay (Auerbach, A.D., et al., 1979, Am. 
J - Hum. Genet. 31 ( 1) : 77-81) . The patient had undergone no 
interventional therapy other than Danazol administration. He 



cells and once with platelets) . 

The source of neonatal blood for the hematopoietic 
reconstitution was a female sibling, who was compatible with 
the patient for HLA and red cell antigens. By study of the 
in utero sibling's fibroblasts obtained at amniocentesis, the 
sibling was found to be a four antigen match. The chromosome 
breakage test (Auerback, A.D., et al, 1979, Am. J. Hum. 
Genet. 31(1):77-81) demonstrated that the female sibling did 
not suffer from Fanconi's anemia. 

Approximately 150 ml neonatal blood was collected from 
the umbilical cord and placenta of the sibling at birth, and 
was diluted 1:1 in sterile pyrogen-free saline containing 
DMSO to a final concentration of 10% DMSO. The blood was 
then shipped under sterile conditions by overnight mail to a 
processing site, where it was frozen slowly, in transplant- 
ation bags, in a time-freezing apparatus, and was stored in 
liquid nitrogen. 

Prior to freezing, a sample of the diluted blood was 
assayed to determine hematopoietic progenitor cell counts as 
described in Section 6.6, supra . The results are shown in 
Table X. 



had undergone transfusions on two occasions (once with red 



25 



30 



35 



i) 



-113- 



TABLE X 

HEMATOPOIETIC PROGENITOR CELLS IN DONOR NEONATAL BLOOD 
5 Before Freezing Cord Blood Placental Blood Total 
Total Nucleated 

Cells 1.05 x 10 9 1.42 x 10 1.2 x 10 9 

Granulocyte- 
Macrophage 
Q Progenitors 

(CFU-GM) 2.23 X 10^ 0.13 X 10° 2.46 x 10 

Erythroid Pro- 
genitors 

(BFU-E) 3.72 X 10 b 0.25 X 10 3.97 X 10 5 

15 Multipotential 
Progenitors 

(CFU-GEMM) 3.57 x 10 0.28 x 10 0.39 X 10 4 



20 



25 



30 



35 



After a test freeze-thaw on a sample of the neonatal 
blood (1 month frozen) , recovery of viable hematopoietic 
progenitor cells was as follows (as assessed by in vitro 
hematopoietic progenitor cell colony assays as described in 
Section 6.6): 100% of CFU-GM, 45% of BFU-E, and 75% of CFU- 
GEMM. 

The patient was conditioned for hematopoietic 
reconstitution by methods similar to those which have been 
used for conditioning nonconstitutional aplastic anemia 
(Gluckman, E., et al . , 1984, in Aplastic Anaemia, Stem Cell 
Biology and Advances in Treatment, Young, N.S., et al., eds. 
Alan R. Liss, Inc., New York, pp. 325-333; incorporated by 
reference herein) except that dosages of chemoradiotherapy 
were decreased. The patient was administered cytoxin® 
(cyclophosphamide) intravenously at a dosage of 5 mg/kg/day 
at six, five, four, and three days prior to neonatal blood 



-114- 



10 



15 



20 



25 



30 



35 



infusion, for a total of 2 0 mg/kg. One day prior to 
infusion, the patient was subjected to thoraco-abdominal 
irradiation with 500 rads and administered Cyclosporin A. 

The frozen blood sample was shipped, under liquid 
nitrogen, to the site of patient treatment, where it was 
thawed in a water bath. Approximately 3 00 ml of the thawed 
blood sample was infused intravenously into the patient for 
treatment of the Fanconi's anemia. 

6.13. FLOWCHART: DESCRIPTION OF A SERVICE 
In a particular embodiment of the invention, the isola- 
tion and preservation of neonatal hematopoietic stem and 
progenitor cells is envisioned as a service offered to each 
prospective cell donor, which can comprise the steps listed 
below. The description is meant for illustrative purposes 
only, in no way limiting the scope of the invention. 

1 . Contact 

Initial contact is made between an expectant 
mother (client) and the obstetrician, who arranges the ser- 
vice. 

2. Blood Collection 

In the obstetrical ward, after the infant has been 
delivered and separated from the cord in the usual way, blood 
is drawn from the cord into a specially designed receptacle, 
which is sealed and placed in a customized shipping con- 
tainer, together with a data-form, completed by a member of 
the obstetrical team, giving details of the birth. 

3 . Transport 

Once daily, an overnight freight carrier collects 
the shipping containers from the obstetrical wards, and 
transports them to processing headquarters by 10:30 A.M. the 
following day. 

4. Registration 





10 



15 



20 



25 



30 



-115- 



Upon receipt at headquarters, each container is 
catalogued . The blood enters the laboratory for processing 
(optional) . 

5. Blood Processing (optional) 

The cells are separated, and the white cells, 
which include the stem and progenitor cells, are retained for 
storage, 

6. Testing 

The separated cells undergo routine testing (see 
Section 5.1.2, supra ) . In exceptional cases, special testing 
may be indicated to determine whether the sample is contamin- 
ated, e.g. , by maternal blood. Samples may be rejected for 
reason of contamination or other causes. 

7. Packaging and Labeling 

Cells from each accepted sample are dispensed into 
standard freezing vials (cryules) and labeled in conventional 
and computer-generated characters. 

The cells of each individual are allocated to four 
cryules, two of which are assigned for storage to one freezer 
and two to another, independently-serviced, freezer. A fifth 
cryule contains cells set aside for testing of identity, 
viability, and function, when withdrawal of cells is required 
for therapy. 

Labels are printed by computer, using a special 
printer, on silk, which withstands immersion in liquid 
nitrogen. The label data include the registration number, in 
machine readable and human readable characters, date of 
freezing, cryule number (1-4, 5) and freezer assignment (A 
and B) . 

8. Freezing and Storage 

The cryules are subjected to slow freezing, and 
assigned to two separately maintained liquid nitrogen refri- 
gerators . 

9 . Permanent Records 



35 






-116- 



The entire preparative history is entered into the 



10 



15 



20 



25 



30 



permanent records, including location within cryostorage 
modules. For example, data input for each donor for main- 
tenance in the computer records can comprise: 

Registration number 

Name 

Sex 

Date of birth 

Place of birth (hospital identification) 

Birth certificate number 

Name of mother 

Date of receipt of cells 

Date of freezing 

Freezer positions 

Obstetrical data 

(a) special circumstances of birth 

(b) if twin, registration number of co-twin 

(c) any health disorder of the mother- 
Test results 

(a) differential cell counts 

(b) bacterial cultures 

(c) other tests performed 

10. Notification to Client 

The client is notified of the registration number, 
for preservation with child's documents, and is asked for 
information not available at the time of birth (given name, 
birth number) , for inclusion in permanent records. 

11. Withdrawal of Cells for Clinical Use 

Requests for cells for treatment of the donor are 
made on behalf of the donor by a suitably accredited physi- 
cian affiliated with an appropriate hospital unit. Cells are 
withdrawn from the cell bank and matched for identity with 
the recipient. The cells are also tested for viability and 
microbial contamination, and quantified in terms of stem 
cell, progenitor cell, and other categories. Further tests 




-117- 

are conducted as required. Cells and an accompanying report 
are delivered to the medical institution designated by the 
physician. An appropriate notation is entered in the perma- 
nent records. 

It is apparent that many modifications and variations of 
this invention as hereinabove set forth may be made without 
departing from the spirit and scope thereof. The specific 
embodiments described are given by way of example only and 
the invention is limited only by the terms of the appended 
claims . 



15 



20 



25 



30 



35 



at